Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Reprod Fertil Dev ; 29(3): 609-620, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26488911

RESUMO

A dynamic partnership between follicle-stimulating hormone (FSH) and activin is required for normal Sertoli cell development and fertility. Disruptions to this partnership trigger Sertoli cells to deviate from their normal developmental pathway, as observed in inhibin α-knockout (Inha-KO) mice, which feature Sertoli cell tumours in adulthood. Here, we identified the developmental windows by which adult Sertoli cell tumourigenesis is most FSH sensitive. FSH was suppressed for 7 days in Inha-KO mice and wild-type littermates during the 1st, 2nd or 4th week after birth and culled in the 5th week to assess the effect on adult Sertoli cell development. Tumour growth was profoundly reduced in adult Inha-KO mice in response to FSH suppression during Weeks 1 and 2, but not Week 4. Proliferative Sertoli cells were markedly reduced in adult Inha-KO mice following FSH suppression during Weeks 1, 2 or 4, resulting in levels similar to those in wild-type mice, with greatest effect observed at the 2 week time point. Apoptotic Sertoli cells increased in adult Inha-KO mice after FSH suppression during Week 4. In conclusion, acute FSH suppression during the 1st or 2nd week after birth in Inha-KO mice profoundly suppresses Sertoli cell tumour progression, probably by inhibiting proliferation in the adult, with early postnatal Sertoli cells being most sensitive to FSH action.


Assuntos
Inibinas/metabolismo , Tumor de Células de Sertoli/patologia , Espermatogênese/genética , Neoplasias Testiculares/patologia , Ativinas/sangue , Animais , Hormônio Foliculoestimulante/sangue , Inibinas/genética , Masculino , Camundongos , Camundongos Knockout , Tumor de Células de Sertoli/genética , Tumor de Células de Sertoli/metabolismo , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Neoplasias Testiculares/genética , Neoplasias Testiculares/metabolismo , Testículo/metabolismo , Testículo/patologia
2.
Nature ; 465(7299): 798-802, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20383121

RESUMO

The ovarian hormones oestrogen and progesterone profoundly influence breast cancer risk, underpinning the benefit of endocrine therapies in the treatment of breast cancer. Modulation of their effects through ovarian ablation or chemoprevention strategies also significantly decreases breast cancer incidence. Conversely, there is an increased risk of breast cancer associated with pregnancy in the short term. The cellular mechanisms underlying these observations, however, are poorly defined. Here we demonstrate that mouse mammary stem cells (MaSCs) are highly responsive to steroid hormone signalling, despite lacking the oestrogen and progesterone receptors. Ovariectomy markedly diminished MaSC number and outgrowth potential in vivo, whereas MaSC activity increased in mice treated with oestrogen plus progesterone. Notably, even three weeks of treatment with the aromatase inhibitor letrozole was sufficient to reduce the MaSC pool. In contrast, pregnancy led to a transient 11-fold increase in MaSC numbers, probably mediated through paracrine signalling from RANK ligand. The augmented MaSC pool indicates a cellular basis for the short-term increase in breast cancer incidence that accompanies pregnancy. These findings further indicate that breast cancer chemoprevention may be achieved, in part, through suppression of MaSC function.


Assuntos
Estrogênios/metabolismo , Glândulas Mamárias Animais/citologia , Progesterona/metabolismo , Células-Tronco/citologia , Animais , Aromatase/metabolismo , Inibidores da Aromatase/farmacologia , Antígeno CD24/metabolismo , Contagem de Células , Receptores ErbB/metabolismo , Estrogênios/farmacologia , Feminino , Humanos , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Letrozol , Camundongos , Nitrilas/farmacologia , Ovariectomia , Comunicação Parácrina/efeitos dos fármacos , Gravidez , Prenhez/fisiologia , Progesterona/antagonistas & inibidores , Progesterona/farmacologia , Ligante RANK/metabolismo , Receptores de Estrogênio/deficiência , Receptores de Progesterona/deficiência , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Triazóis/farmacologia
3.
Reprod Fertil Dev ; 2016 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-26946412

RESUMO

Although oestrogens are essential for spermatogenesis and their biosynthesis is dependent on aromatase expression, the molecular mechanism of aromatase regulation is poorly understood. Our laboratory has demonstrated that liver kinase B1 (LKB1) is a negative regulator of aromatase in the breast by phosphorylating AMP-activated protein kinase (AMPK) and inhibiting the nuclear translocation of the cAMP response element-binding protein-regulated transcription co-activator (CRTC) 2. The aim of this study was to determine the location of testis-associated proteins in the LKB1-CRTC pathway. Aromatase, LKB1, phosphorylated AMPK (pAMPK) and CRTC1-3 were examined by selected immunofluorescent antibodies in testis samples from a prepubertal boy and three fertile men. Aromatase, pAMPK and LKB1 proteins were present in the seminiferous epithelium and interstitium of the testis and were expressed in a differential and developmental manner in particular cell types. The expression pattern of LKB1 was similar to that of pAMPK and inversely related to aromatase expression. CRTC1 and CRTC3 were localised in the seminiferous epithelium, whereas CRTC2 was barely detectable in testis. These results lead to the conclusion that LKB1 is involved in the molecular pathway that underpins aromatase regulation in the testis via CRTC1 and CRTC3 and may be important for the oestrogen-mediated development of germ cells.

4.
Breast Cancer Res ; 15(2): R30, 2013 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-23566437

RESUMO

INTRODUCTION: The majority of postmenopausal breast cancers are estrogen-dependent. Tumor-derived factors, such as prostaglandin E2 (PGE2), stimulate CREB1 binding to cAMP response elements (CREs) on aromatase promoter II (PII), leading to the increased expression of aromatase and biosynthesis of estrogens within human breast adipose stromal cells (ASCs). Hypoxia inducible factor-1α (HIF-1α), a key mediator of cellular adaptation to low oxygen levels, is emerging as a novel prognostic marker in breast cancer. We have identified the presence of a consensus HIF-1α binding motif overlapping with the proximal CRE of aromatase PII. However, the regulation of aromatase expression by HIF-1α in breast cancer has not been characterized. This study aimed to characterize the role of HIF-1α in the activation of aromatase PII. METHODS: HIF-1α expression and localization were examined in human breast ASCs using quantitative PCR (QPCR), Western blotting, immunofluorescence and high content screening. QPCR and tritiated water-release assays were performed to assess the effect of HIF-1α on aromatase expression and activity. Reporter assays and chromatin immunoprecipitation (ChIP) were performed to assess the effect of HIF-1α on PII activity and binding. Treatments included PGE2 or DMOG ((dimethyloxalglycine), HIF-1α stabilizer). Double immunohistochemistry for HIF-1α and aromatase was performed on tissues obtained from breast cancer and cancer-free patients. RESULTS: Results indicate that PGE2 increases HIF-1α transcript and protein expression, nuclear localization and binding to aromatase PII in human breast ASCs. Results also demonstrate that HIF-1α significantly increases PII activity, and aromatase transcript expression and activity, in the presence of DMOG and/or PGE2, and that HIF-1α and CREB1 act co-operatively on PII. There is a significant increase in HIF-1α positive ASCs in breast cancer patients compared to cancer-free women, and a positive association between HIF-1α and aromatase expression. CONCLUSIONS: This study is the first to identify HIF-1α as a modulator of PII-driven aromatase expression in human breast tumor-associated stroma and provides a novel mechanism for estrogen regulation in obesity-related, post-menopausal breast cancer. Together with our on-going studies on the role of AMP-activated protein kinase (AMPK) in the regulation of breast aromatase, this work provides another link between disregulated metabolism and breast cancer.


Assuntos
Tecido Adiposo/metabolismo , Aromatase/genética , Neoplasias da Mama/metabolismo , Dinoprostona/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Estromais/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/patologia , Aromatase/metabolismo , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/tratamento farmacológico , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Imunoprecipitação da Cromatina , Feminino , Imunofluorescência , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Técnicas Imunoenzimáticas , Ocitócicos/farmacologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Células Estromais/patologia , Células Tumorais Cultivadas
5.
Breast Cancer Res Treat ; 138(1): 193-203, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23338760

RESUMO

Expression of the oestrogen producing enzyme, aromatase, is regulated in a tissue-specific manner by its encoding gene CYP19A1. In post-menopausal women, the major site for oestrogen production in the breast is the adipose, where CYP19A1 transcription is driven by the distal promoter I.4 (PI.4). Transcripts via this promoter are also elevated in breast adipose fibroblasts (BAFs) adjacent to a tumour. PI.4 expression is stimulated by a number of cytokines, and TNFα is one such factor. The transcriptional mechanisms induced by TNFα to stimulate PI.4 are poorly characterised. We show that the early growth response (Egr) transcription factors play an important role in the TNFα-induced signalling pathway resulting in elevated PI.4 transcription. TNFα treatment of BAFs increases mRNA levels of all four Egr family members, with EGR2 being the most highly expressed. Overexpression of EGR2 causes an increase in endogenous CYP19A1 expression in preadipocyte Simpson-Golabi-Behmel syndrome cells, driven by increases in PI.4-specific transcripts. PI.4 luciferase reporter activity is increased in a dose-dependent manner by EGR2, EGR3 and EGR4, with EGR2 showing the most potent activation of promoter activity. Deletion analysis indicates that this promoter activity is being indirectly mediated by a short region of the promoter not containing any previously characterised binding sites, and we further show that EGR2 does not bind directly or indirectly to this promoter region. However, siRNA knockdown of the Egrs reduces the total and PI.4-derived CYP19A1 transcription in BAFs. These studies unveil a novel component of the aromatase gene regulatory network and further enhance the complexity of oestrogen production in the breast.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Aromatase/genética , Mama/citologia , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Animais , Linhagem Celular , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Feminino , Expressão Gênica , Genes Reporter , Humanos , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , RNA Mensageiro/genética , Elementos de Resposta , Deleção de Sequência , Ativação Transcricional
6.
Breast Cancer Res Treat ; 142(1): 211-23, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24122391

RESUMO

The interaction between breast tumor epithelial and stromal cells is vital for initial and recurrent tumor growth. While breast cancer-associated stromal cells provide a favorable environment for proliferation and metastasis, the molecular mechanisms contributing to this process are not fully understood. Nuclear receptors (NRs) are intracellular transcription factors that directly regulate gene expression. Little is known about the status of NRs in cancer-associated stroma. Nuclear Receptor Low-Density Taqman Arrays were used to compare the gene expression profiles of all 48 NR family members in a collection of primary cultured cancer-associated fibroblasts (CAFs) obtained from estrogen receptor (ER)α positive breast cancers (n = 9) and normal breast adipose fibroblasts (NAFs) (n = 7). Thirty-three of 48 NRs were expressed in both the groups, while 11 NRs were not detected in either. Three NRs (dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1 (DAX-1); estrogen-related receptor beta (ERR-ß); and RAR-related orphan receptor beta (ROR-ß)) were only detected in NAFs, while one NR (liver receptor homolog-1 (LRH-1)) was unique to CAFs. Of the NRs co-expressed, four were significantly down-regulated in CAFs compared with NAFs (RAR-related orphan receptor-α (ROR-α); Thyroid hormone receptor-ß (TR-ß); vitamin D receptor (VDR); and peroxisome proliferator-activated receptor-γ (PPAR-γ)). Quantitative immunohistochemistry for LRH-1, TR-ß, and PPAR-γ proteins in stromal fibroblasts from an independent panel of breast cancers (ER-positive (n = 15), ER-negative (n = 15), normal (n = 14)) positively correlated with mRNA expression profiles. The differentially expressed NRs identified in tumor stroma are key mediators in aromatase regulation and subsequent estrogen production. Our findings reveal a distinct pattern of NR expression that therefore fits with a sustained and increased local estrogen microenvironment in ER-positive tumors. NRs in CAFs may provide a new avenue for the development of intratumoral-targeted therapies in breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Células Estromais/metabolismo , Idoso , Idoso de 80 Anos ou mais , Neoplasias da Mama/genética , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Gradação de Tumores , Receptor ErbB-2/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Fatores de Risco
7.
Am J Physiol Endocrinol Metab ; 302(5): E576-84, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22185842

RESUMO

The aim of this study was to create a comprehensive mouse model of the metabolic syndrome by crossing aromatase-deficient (ArKO) mice with apolipoprotein E-deficient (ApoE(-/-)) mice. Successive crossbreeding of ArKO with ApoE(-/-)-deficient mice generated double knockout, MetS-Tg mice. The phenotypic characteristics of the MetS-Tg mice were assessed at 3, 6, and 12 mo of age and compared with age- and sex-matched wild-type (WT) controls. Blood pressure and heart rate were recorded by a noninvasive, computerized tail-cuff system. Oral glucose and intraperitoneal insulin tolerance tests were performed. Serum cholesterol levels were measured by a combined quantitative colorimetric assay. Plasma adiponectin, C-reactive protein (CRP), insulin, interleukin-6 (IL-6), leptin, resistin, and tumor necrosis factor-α (TNF-α) were measured by multiplexed ELISA. MetS-Tg mice displayed significantly increased body weight, central obesity, and elevated blood pressure at all three ages compared with WT mice. Elevated serum cholesterol was associated with higher triglycerides and LDL/VLDL cholesterol particles and was accompanied by a decrease in HDL and histological evidence of fatty liver. MetS-Tg mice of all ages showed impaired glucose tolerance. At 12 mo, MetS-Tg mice had elevated plasma levels of CRP, IL-6, leptin, and TNF-α, but resistin levels were largely unchanged. We now report that this combination of gene knockouts produces a novel strain of mice that display the diverse clinical features of the metabolic syndrome, including central obesity, progressive hypertension, an adverse serum lipid profile, fatty liver, glucose intolerance, insulin resistance, and evidence of an inflammatory state.


Assuntos
Apolipoproteínas E/fisiologia , Aromatase/fisiologia , Modelos Animais de Doenças , Síndrome Metabólica/fisiopatologia , Tecido Adiposo Branco/imunologia , Tecido Adiposo Branco/patologia , Animais , Aorta/imunologia , Aorta/patologia , Apolipoproteínas E/genética , Aromatase/genética , Aterosclerose/etiologia , Proteína C-Reativa/análise , Cruzamentos Genéticos , Citocinas/sangue , Fígado Gorduroso/etiologia , Feminino , Intolerância à Glucose/etiologia , Hiperlipidemias/etiologia , Hipertensão/etiologia , Resistência à Insulina , Fígado/imunologia , Fígado/patologia , Masculino , Síndrome Metabólica/sangue , Síndrome Metabólica/imunologia , Síndrome Metabólica/patologia , Camundongos , Camundongos Knockout , Obesidade Abdominal/etiologia
8.
Breast Cancer Res Treat ; 132(2): 765-71, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22237979

RESUMO

The main biological active substance secreted by the pineal gland, melatonin (MLT), counteracts the effects of estrogens in breast cancer via exerting a number of its own oncostatic properties. Recent studies of postmenopausal women have identified that the major metabolite of MLT is statistically significantly associated with a lower risk of developing breast cancer. While MLT production decreases with age, breast cancer risk, however, increases with age and obesity. We hypothesize that MLT inhibits estrogen production in breast adipose fibroblasts (BAFs), the main local source of estrogen in breast tumors of postmenopausal women, by inhibiting transcription of the CYP19A1 gene that encodes the key enzyme aromatase. Normal BAFs were cultured from women undergoing breast reduction surgery, while breast cancer-associated fibroblasts (CAFs) were isolated from three women with estrogen receptor (ER) positive invasive ductal carcinomas. MTNR1A and MTNR1B receptor expression and CYP19A1 mRNA expression following MLT treatments were determined by qRT-PCR. BAFs express the G-protein coupled MLT receptors MTNR1A and MTNR1B with elevated levels of MTNR1A found in CAFs. Treatment of BAFs and CAFs with MLT resulted in significant suppression of CYP19A1 transcription and aromatase activity at pharmacological, physiological and sub-physiological concentrations. MLT suppression occurred through promoter-specific PI.4-, PI.3- and PII-derived CYP19A1 mRNA. Stimulation of CYP19A1 PII-mRNA and aromatase activity by prostaglandin E(2) (PGE(2)) were significantly attenuated by physiological doses of MLT. Lower levels of MLT in aging women may increase the risk of progressing ER-positive breast cancer through a decreased ability to suppress CYP19A1 expression and subsequent local estrogen production in BAFs/CAFs.


Assuntos
Tecido Adiposo/enzimologia , Aromatase/metabolismo , Neoplasias da Mama/enzimologia , Carcinoma Ductal de Mama/enzimologia , Fibroblastos/enzimologia , Melatonina/metabolismo , Tecido Adiposo/patologia , Aromatase/genética , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Células Cultivadas , Dinoprostona/metabolismo , Regulação para Baixo , Feminino , Fibroblastos/patologia , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Receptor MT1 de Melatonina/metabolismo , Receptor MT2 de Melatonina/metabolismo , Receptores de Estrogênio/metabolismo , Transcrição Gênica
9.
Transgenic Res ; 21(2): 415-28, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21614586

RESUMO

Aromatase converts androgens to estrogens and it is expressed in gonads and non-reproductive tissues (e.g. brain and adipose tissues). As circulating levels of estrogens in males are low, we hypothesize that local estrogen production is important for the regulation of physiological functions (e.g. metabolism) and pathological development (e.g. breast and prostate cancers) by acting in a paracrine and/or intracrine manner. We generated a tissue-specific doxycycline-inducible, aromatase transgenic mouse to test this hypothesis. The transgene construct (pTetOAROM) consists of a full-length human aromatase cDNA (hAROM) and a luciferase gene under the control of a bi-directional tetracycline-responsive promoter (pTetO), which is regulated by transactivators (rtTA or tTA) and doxycycline. Our in vitro studies using MBA-MB-231tet cells stably expressing rtTA, showed that doxycycline treatment induced transgene expression of hAROM transcripts by 17-fold (P = 0.01), aromatase activity by 26-fold, (P = 0.0008) and luciferase activity by 9.6-fold (P = 0.0006). Pronuclear microinjection of the transgene generated four pTetOAROM founder mice. A male founder was bred with a female mammary gland-specific rtTA mouse (MMTVrtTA) to produce MMTVrtTA-pTetOAROM double-transgenic mice. Upon doxycycline treatment via drinking water, human aromatase expression was detected by RT-PCR, specifically in mammary glands, salivary glands and seminal vesicles of double-stransgenic mice. Luciferase expression and activity was detected in these tissues by in vivo bioluminescence imaging, in vitro luciferase assay and RT-PCR. In summary, we generated a transgenic mouse model that expresses the human aromatase transgene in a temporal- and spatial-specific manner, which will be a useful model to study the physiological importance of local estrogen production.


Assuntos
Aromatase/metabolismo , Doxiciclina/farmacologia , Regulação Enzimológica da Expressão Gênica , Animais , Aromatase/genética , Linhagem Celular Tumoral , Clonagem Molecular , DNA Complementar/genética , DNA Complementar/metabolismo , Doxiciclina/administração & dosagem , Ativação Enzimática , Ensaios Enzimáticos , Feminino , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Humanos , Luciferases de Vaga-Lume/genética , Luciferases de Vaga-Lume/metabolismo , Medições Luminescentes/métodos , Masculino , Glândulas Mamárias Humanas/citologia , Glândulas Mamárias Humanas/metabolismo , Camundongos , Camundongos Transgênicos , Microinjeções , Plasmídeos/genética , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Glândulas Salivares/citologia , Glândulas Salivares/metabolismo , Glândulas Seminais/citologia , Glândulas Seminais/metabolismo , Transgenes
10.
Breast Cancer Res Treat ; 123(2): 591-6, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20300828

RESUMO

AMP-activated protein kinase (AMPK) is recognized as a master regulator of energy homeostasis. In concert with the AMPK-kinase LKB1, it has been shown to provide a molecular link between obesity and postmenopausal breast cancer via its actions to inhibit aromatase expression, hence estrogen production, within the breast. The anti-diabetic drug metformin is known to increase the activity of AMPK and was therefore hypothesized to inhibit aromatase expression in primary human breast adipose stromal cells. Results demonstrate that metformin significantly decreases the forskolin/phorbol ester (FSK/PMA)-induced expression of aromatase at concentrations of 10 and 50 muM. Consistent with the hypothesized actions of metformin to increase AMPK activity, treatment with 50 muM metformin results in a significant increase in phosphorylation of AMPK at Thr172. Interestingly, metformin also causes a significant increase in LKB1 protein expression and promoter activity, thereby providing for the first time an additional mechanism by which metformin activates AMPK. Furthermore, metformin inhibits the nuclear translocation of CRTC2, a CREB-coactivator known to increase aromatase expression which is also a direct downstream target of AMPK. Overall, these results suggest that metformin would reduce the local production of estrogens within the breast thereby providing a new key therapeutic tool that could be used in the neoadjuvant and adjuvant settings and conceivably also as a preventative measure in obese women.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Tecido Adiposo/efeitos dos fármacos , Inibidores da Aromatase/farmacologia , Aromatase/metabolismo , Mama/efeitos dos fármacos , Metformina/farmacologia , Células Estromais/efeitos dos fármacos , Quinases Proteína-Quinases Ativadas por AMP , Transporte Ativo do Núcleo Celular , Tecido Adiposo/enzimologia , Animais , Aromatase/genética , Sítios de Ligação , Mama/enzimologia , Células COS , Chlorocebus aethiops , Colforsina/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Feminino , Humanos , Fosforilação , Regiões Promotoras Genéticas , Proteínas Serina-Treonina Quinases/metabolismo , Células Estromais/enzimologia , Acetato de Tetradecanoilforbol/farmacologia , Treonina , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
11.
Clin Exp Pharmacol Physiol ; 37(1): 78-82, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19566833

RESUMO

1. Oestrogen plays an important role in ageing and ageing-related development. Lack of oestrogen prompts endocrine cell ageing of the ovary, whereas oestrogen overflow impacts on epithelial cell neoplastic development. 2. Recent studies indicate that oestrogen regulates cell proliferative fates by a mechanism of reprogramming the size of telomeres (ends of chromosomes) in the oestrogen target cells. This is achieved by upregulating the telomerase reverse transcriptase (TERT) gene in a temporal and spatial manner. 3. Currently, the relationship between oestrogen and telomerase activity in regulating productive cell development and function remains elusive. A number of lines of evidence suggest that telomerase is a downstream target of oestrogen in oestrogen-dependent reproductive ageing and neoplastic development. 4. The present minireview discusses our current understanding of the mechanisms by which telomerase maintenance of telomere homeostasis mediates oestrogen-induced ageing and tumourigenesis in the ovary under physiological and pathological conditions.


Assuntos
Envelhecimento/fisiologia , Estrogênios/fisiologia , Regulação Neoplásica da Expressão Gênica , Neoplasias/enzimologia , Neoplasias Ovarianas/enzimologia , Ovário/fisiologia , Telomerase/fisiologia , Telômero/enzimologia , Envelhecimento/genética , Senescência Celular/genética , Feminino , Humanos , Modelos Biológicos , Neoplasias/genética , Neoplasias/fisiopatologia , Neoplasias Ovarianas/fisiopatologia , Ovário/patologia , Telomerase/genética
12.
Mol Cell Neurosci ; 41(1): 1-7, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19185610

RESUMO

The aromatase knockout (ArKO) mouse is completely estrogen deficient. We previously detected apoptosis in the hypothalamus of 1 year-old male ArKO mice. This study shows that 12 week-old female ArKO mice display spontaneous apoptosis of pyramidal neurons in the frontal cortex while wild-type (WT) littermates show no signs of apoptosis. Concomitantly, bcl-2 related anti-apoptotic genes are down-regulated whereas the pro-apoptotic gene TRADD is up-regulated in the female ArKO frontal cortex. This phenotype can be rescued by 3-week replacement of 17beta-estradiol. Furthermore, the apoptosis phenotype is exacerbated in 12-15 month-old female ArKO mice, which have 30% less neurons in the frontal cortex and lower brain weights than WT counterparts. These data show that estrogens are essential for the survival of female cortical neurons even in the absence of pathological conditions or external assaults. Our observations also demonstrate the sexually dimorphic susceptibility of neurons to estrogen deficiency.


Assuntos
Apoptose/fisiologia , Aromatase , Estrogênios/deficiência , Lobo Frontal/metabolismo , Lobo Frontal/patologia , Animais , Aromatase/genética , Aromatase/metabolismo , Caspase 3/metabolismo , Sobrevivência Celular , Proteínas de Ligação a DNA , Estradiol/administração & dosagem , Feminino , Lobo Frontal/citologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neurônios/patologia , Proteínas Nucleares/metabolismo , Tamanho do Órgão
13.
Endokrynol Pol ; 61(1): 126-34, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20205115

RESUMO

Aromatase is a member of the cytochrome P450 superfamily that catalyzes the conversion of androgens (C(19)), namely testosterone and androstenedione, into oestrogens (C(18)), oestradiol, and oestrone, respectively. The enzyme is active in various tissues in both females and males, thus oestrogens are produced not only in gonads but also in extra-gonadal localizations such as brain, adipose tissue, breast, skin, and bone. Aromatase gene CYP19A1 located on chromosome 15 comprises nine coding exons and a number of alternative non-coding first exons that regulate tissue-specific expression. Studies on local regulation of aromatase expression and activity are important for understanding processes such as growth of oestrogen-dependent breast cancer. Rare clinical conditions of aromatase deficiency and excess have revealed some new and unexpected oestrogen functions in metabolism and bone health in both women and men. They were further studied using transgenic animal models such as aromatase knockout mice (ArKO) or (AROM+) mice overexpressing human aromatase. Research on aromatase was important for its practical outcome as it contributed to the development of aromatase inhibitors (AIs), an effective and safe group of drugs for the first-line endocrine therapy of breast cancer. Further studies are needed to establish AIs application in other oestrogen-dependent conditions, to overcome the resistance in breast cancer patients, and to develop tissue-specific selective inhibitors. (Pol J Endocrinol 2010; 61 (1): 126-134).


Assuntos
Aromatase/metabolismo , Animais , Aromatase/genética , Inibidores da Aromatase/uso terapêutico , Osso e Ossos/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Estrogênios/biossíntese , Feminino , Humanos , Masculino , Camundongos , Polimorfismo Genético
14.
BMC Cancer ; 9: 148, 2009 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-19445691

RESUMO

BACKGROUND: Until recently, the molecular mechanisms explaining increased incidence of ovarian and breast cancers in carriers of BRCA1 gene mutations had not been clearly understood. Of significance is the finding that BRCA1 negatively regulates aromatase expression in vitro. Our objective was to characterise aromatase gene (CYP19A1) and its promoter expression in breast adipose and ovarian tissue in BRCA1 mutation carriers and unaffected controls. METHODS: We measured aromatase transcripts, total and promoter-specific (PII, PI.3, PI.4) in prophylactic oophorectomy or mastectomy, therapeutic mastectomy, ovarian and breast tissue from unaffected women. RESULTS: We demonstrate that the lack of functional BRCA1 protein correlates to higher aromatase levels in 85% of BRCA1 mutation carriers. This increase is mediated by aberrant transcriptional regulation of aromatase; in breast adipose by increases in promoter II/I.3 and I.4-specific transcripts; and in the ovary with elevation in promoter I.3 and II-specific transcripts. CONCLUSION: Understanding the link between BRCA1 and aromatase is significant in terms of understanding why carcinogenesis is restricted to estrogen-producing tissues in BRCA1 mutation carriers.


Assuntos
Aromatase/genética , Proteína BRCA1/genética , Expressão Gênica , Heterozigoto , Mutação , Adipócitos/metabolismo , Adulto , Aromatase/metabolismo , Austrália , Proteína BRCA1/metabolismo , Mama/metabolismo , Feminino , Humanos , Ovário/metabolismo
15.
Compr Physiol ; 9(2): 457-475, 2019 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-30873596

RESUMO

As adipose tissue depots are active endocrine organs, they secrete a variety of hormones (including estrogens from white adipose) and inflammatory mediators, which have important implications in numerous obesity-associated diseases. Adipose tissues are broadly characterized as consisting of white, beige, and brown depot types. The endocrine, metabolic, and inflammatory profiles of adipose are depot dependent and influenced by the estrogenic and androgenic status of the adipose tissue. Estrogen receptors mediate both the genomic and nongenomic actions of estrogens and are expressed in the brain, heart, and other peripheral tissues. All three known estrogen receptor α (ERα) and estrogen receptor ß (ERß), and the G-protein coupled estrogen receptor (GPER/GPR30) are expressed in white adipose and can modulate adipose mass. Expression of each receptor is dependent on depot location, adipose cell type, and estrogen levels. Estrogen receptor expression profiles in beige and brown adipocytes are less well established. This review will discuss the effects of estrogens on the differential deposition of the major adipose tissues and the impact of estrogens within white adipose depots. © 2019 American Physiological Society. Compr Physiol 9:457-475, 2019.


Assuntos
Tecido Adiposo/metabolismo , Estrogênios/metabolismo , Adiposidade , Animais , Neoplasias da Mama/metabolismo , Feminino , Humanos , Glândulas Mamárias Humanas/metabolismo
16.
Breast Cancer Res Treat ; 112(3): 429-35, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18181018

RESUMO

Aromatase is a member of the cytochrome P450 superfamily of enzymes which catalyses the rate-limiting step in the biosynthesis of estrogens. A number of clinical studies have highlighted the importance of local estrogen production in adipose tissue. In particular, in the postmenopausal woman, the degree of her estrogenization is mainly determined by the extent of her adiposity and it is this extragonadal source of estrogen that likely contributes to breast cancer development and progression. The mechanisms regulating aromatase expression in adipose tissue however, have not been fully elucidated. In this study, we have characterised the expression of aromatase and its activity in a human preadipocyte cell strain, SGBS. Aromatase is expressed in SGBS cells and its expression and activity are strongly stimulated by forskolin (FSK) and phorbol 12-myristate-13-acetate (PMA) treatment. Consistent with this, FSK and PMA treatment also increased activation of the proximal aromatase promoter, promoter II. These findings mimic those that have previously been shown in isolated primary human preadipocytes. These data suggest that SGBS cells are a valuable model with which to further elucidate the mechanisms regulating aromatase expression, and therefore local estrogen synthesis in human adipose tissue.


Assuntos
Adipócitos/enzimologia , Aromatase/biossíntese , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Aromatase/metabolismo , Linhagem Celular , Colforsina/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , DNA Complementar/metabolismo , Progressão da Doença , Estrogênios/metabolismo , Humanos , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acetato de Tetradecanoilforbol/farmacologia , Fatores de Tempo
17.
Adv Exp Med Biol ; 630: 112-32, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18637488

RESUMO

Estrogen has been positively linked to the pathogenesis and growth of three common women's cancers (breast, endometrium and ovary). A single gene encodes the key enzyme for estrogen biosynthesis named aromatase, inhibition of which effectively eliminates estrogen production in the entire body. Aromatase inhibitors successfully treat breast cancer, whereas their roles in endometrial and ovarian cancers are less dear. Ovary, testis, adipose tissue, skin, hypothalamus and placenta express aromatase normally, whereas breast, endometrial and ovarian cancers overexpress aromatase and produce local estrogen exerting paracrine and intracrine effects. Tissue specific promoters distributed over a 93 kilobase regulatory region upstream of a common coding region alternatively control aromatase expression. A distinct set of transcription factors regulates each promoter in a signaling pathway- and tissue-specific manner. In cancers ofbreast, endometrium and ovary, aromatase expression is primarly regulated by increased activity of the proximally located promoter 1.3/II region. Promoters I.3 and II lie 215 bp from each other and are coordinately stimulated by PGE2 via a cAMP-PKA-dependent pathway. In breast adipose fibroblasts exposed to PGE2 secreted by malignant epithelial cells, activation of PKC potentiates cAMP-PKA-dependent induction ofaromatase. Thus, inflammatory substances such as PGE2 may play important roles in inducing local production of estrogen that promotes tumor growth.


Assuntos
Aromatase/genética , Expressão Gênica , Neoplasias/genética , Tecido Adiposo/fisiologia , Inibidores da Aromatase/uso terapêutico , Endometriose/complicações , Feminino , Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Hormônios/farmacologia , Humanos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/etiologia
18.
Endocrinology ; 148(2): 566-74, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17068134

RESUMO

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.


Assuntos
Receptor beta de Estrogênio/metabolismo , Próstata/fisiopatologia , Hiperplasia Prostática/fisiopatologia , Animais , Animais Recém-Nascidos , Aromatase/deficiência , Proliferação de Células , Epitélio/metabolismo , Epitélio/transplante , Receptor beta de Estrogênio/agonistas , Estrogênios/deficiência , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID , Próstata/metabolismo , Próstata/patologia , Hiperplasia Prostática/etiologia , Hiperplasia Prostática/metabolismo , Hiperplasia Prostática/patologia , Receptores de Superfície Celular/metabolismo , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Transdução de Sinais , Células Estromais/metabolismo , Células Estromais/transplante , Distribuição Tecidual , Transplante Heterotópico
19.
Biol Psychiatry ; 61(3): 359-66, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-16566897

RESUMO

BACKGROUND: Aromatase converts androgen to estrogen. Thus, the aromatase knockout (ArKO) mouse is estrogen deficient. We investigated the compulsive behaviors of these animals and the protein levels of catechol-O-methyltransferase (COMT) in frontal cortex, hypothalamus and liver. METHODS: Grooming was analyzed during the 20-min period immediately following a water-mist spray. Running wheel activity over two consecutive nights and barbering were analyzed. COMT protein levels were measured by Western analysis. RESULTS: Six-month old male but not female ArKO mice develop compulsive behaviors such as excessive barbering, grooming and wheel-running. Excessive activities were reversed by 3 weeks of 17beta-estradiol replacement. Interestingly, the presentation of compulsive behaviors is accompanied by concomitant decreases (p < .05) in hypothalamic COMT protein levels in male ArKO mice. These values returned to normal upon 17beta-estradiol treatment. In contrast, hepatic and frontal cortex COMT levels were not affected by the estrogen status, indicating region- and tissue-specific regulation of COMT levels by estrogen. No differences in COMT levels were detectable between female animals of both genotypes. CONCLUSIONS: This study describes the novel observation of a possible link between estrogen, COMT and development of compulsive behaviors in male animals which may have therapeutic implications in obsessive compulsive disorder (OCD) patients.


Assuntos
Comportamento Compulsivo/fisiopatologia , Comportamento Compulsivo/psicologia , Estrogênios/deficiência , Animais , Aromatase/genética , Aromatase/fisiologia , Western Blotting , Encéfalo/enzimologia , Catecol O-Metiltransferase/genética , Catecol O-Metiltransferase/metabolismo , Comportamento Compulsivo/genética , Estradiol/farmacologia , Estrogênios/farmacologia , Feminino , Asseio Animal/fisiologia , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia
20.
Behav Neurosci ; 121(2): 356-61, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17469925

RESUMO

Aromatization of testosterone (T) to estradiol (E2) during perinatal development in male rodents plays a significant role in sexual differentiation of the brain and adult behaviors. Exposure to estrogens during development can enhance masculine behaviors in adult females and reduce expression of female-typical behaviors in adult males. Previous studies have shown that, in addition to naturally occurring estrogens, dietary phytoestrogens can affect sexual differentiation. To distinguish between the effects of endogenous T-derived E2 and exogenous phytoestrogens, the authors used an aromatase knockout (ArKO) mouse that cannot produce E2 but is responsive to E2 via estrogen receptors alpha and beta. Dams and their litters were maintained either on a standard mouse chow that was rich in phytoestrogens or on a chow nearly devoid of phytoestrogens. Mice were maintained on their perinatal diets after weaning. Adults of both sexes were gonadectomized and tested for lordosis behavior. In the ArKO females raised on a diet high in phytoestrogens, lordosis was reduced in comparison with females of both genotypes on the low phytoestrogen diet. The authors' findings suggest that dietary phytoestrogen consumption may partially defeminize adult female sexual behavior in the mouse.


Assuntos
Aromatase/genética , Fitoestrógenos/farmacologia , Comportamento Sexual Animal/efeitos dos fármacos , Animais , Aromatase/fisiologia , Dieta , Estradiol/fisiologia , Feminino , Hipotálamo/fisiologia , Camundongos , Camundongos Knockout , Orquiectomia , Ovariectomia , Postura/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA