Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
FASEB J ; 35(5): e21536, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33913559

RESUMO

Obesity is a risk factor for postmenopausal breast cancer. Obesity-related inflammation upregulates aromatase expression, the rate-limiting enzyme for estrogen synthesis, in breast adipose tissue (BAT), increasing estrogen production and cancer risk. The regulation of aromatase gene (CYP19A1) in BAT is complex, and the mechanisms linking obesity and aromatase dysregulation are not fully understood. An obesity-associated factor that could regulate aromatase is the CC chemokine ligand (CCL) 2, a pro-inflammatory factor that also activates signaling pathways implicated in CYP19A1 transcription. By using human primary breast adipose stromal cells (ASCs) and aromatase reporter (hARO-Luc) mouse mammary adipose explants, we demonstrated that CCL2 enhances the glucocorticoid-mediated CYP19A1 transcription. The potential mechanism involves the activation of PI.4 via ERK1/2 pathway. We also showed that CCL2 contributes to the pro-inflammatory milieu and aromatase expression in obesity, evidenced by increased expression of CCL2 and CYP19A1 in mammary tissues from obese hARO-Luc mice, and subcutaneous adipose tissue from obese women. In summary, our results indicate that postmenopausal obesity may promote CCL2 production in BAT, leading to exacerbation of the menopause-related inflammatory state and further stimulation of local aromatase and estrogens. These results provide new insights into the regulation of aromatase and may aid in finding approaches to prevent breast cancer.


Assuntos
Aromatase/metabolismo , Mama/metabolismo , Quimiocina CCL2/metabolismo , Regulação Enzimológica da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Obesidade/fisiopatologia , Ativação Transcricional , Animais , Aromatase/genética , Mama/citologia , Quimiocina CCL2/genética , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos
2.
Am J Pathol ; 188(12): 2890-2901, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30273606

RESUMO

The role of adrenal androgens as drivers for castration-resistant prostate cancer (CRPC) growth in humans is generally accepted; however, the value of preclinical mouse models of CRPC is debatable, because mouse adrenals do not produce steroids activating the androgen receptor. In this study, we confirmed the expression of enzymes essential for de novo synthesis of androgens in mouse adrenals, with high intratissue concentration of progesterone (P4) and moderate levels of androgens, such as androstenedione, testosterone, and dihydrotestosterone, in the adrenal glands of both intact and orchectomized (ORX) mice. ORX alone had no effect on serum P4 concentration, whereas orchectomized and adrenalectomized (ORX + ADX) resulted in a significant decrease in serum P4 and in a further reduction in the low levels of serum androgens (androstenedione, testosterone, and dihydrotestosterone), measured by mass spectrometry. In line with this, the serum prostate-specific antigen and growth of VCaP xenografts in mice after ORX + ADX were markedly reduced compared with ORX alone, and the growth difference was not abolished by a glucocorticoid treatment. Moreover, ORX + ADX altered the androgen-dependent gene expression in the tumors, similar to that recently shown for the enzalutamide treatment. These data indicate that in contrast to the current view, and similar to humans, mouse adrenals synthesize significant amounts of steroids that contribute to the androgen receptor-dependent growth of CRPC.


Assuntos
Glândulas Suprarrenais/patologia , Adrenalectomia , Androgênios/metabolismo , Modelos Animais de Doenças , Orquiectomia , Neoplasias de Próstata Resistentes à Castração/patologia , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/cirurgia , Animais , Xenoenxertos , Humanos , Masculino , Camundongos , Neoplasias de Próstata Resistentes à Castração/etiologia , Neoplasias de Próstata Resistentes à Castração/metabolismo
3.
Am J Pathol ; 188(1): 216-228, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29126837

RESUMO

The development of castration-resistant prostate cancer (CRPC) is associated with the activation of intratumoral androgen biosynthesis and an increase in androgen receptor (AR) expression. We recently demonstrated that, similarly to the clinical CRPC, orthotopically grown castration-resistant VCaP (CR-VCaP) xenografts express high levels of AR and retain intratumoral androgen concentrations similar to tumors grown in intact mice. Herein, we show that antiandrogen treatment (enzalutamide or ARN-509) significantly reduced (10-fold, P < 0.01) intratumoral testosterone and dihydrotestosterone concentrations in the CR-VCaP tumors, indicating that the reduction in intratumoral androgens is a novel mechanism by which antiandrogens mediate their effects in CRPC. Antiandrogen treatment also altered the expression of multiple enzymes potentially involved in steroid metabolism. Identical to clinical CRPC, the expression levels of the full-length AR (twofold, P < 0.05) and the AR splice variants 1 (threefold, P < 0.05) and 7 (threefold, P < 0.01) were further increased in the antiandrogen-treated tumors. Nonsignificant effects were observed in the expression of certain classic androgen-regulated genes, such as TMPRSS2 and KLK3, despite the low levels of testosterone and dihydrotestosterone. However, other genes recently identified to be highly sensitive to androgen-regulated AR action, such as NOV and ST6GalNAc1, were markedly altered, which indicated reduced androgen action. Taken together, the data indicate that, besides blocking AR, antiandrogens modify androgen signaling in CR-VCaP xenografts at multiple levels.


Assuntos
Antagonistas de Androgênios/farmacologia , Androgênios/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Receptores Androgênicos/metabolismo , Animais , Benzamidas , Linhagem Celular Tumoral , Di-Hidrotestosterona/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Nitrilas , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Testosterona/metabolismo , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
FASEB J ; 32(6): 3361-3370, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29401621

RESUMO

White adipose tissue inflammation is linked with increased aromatase gene expression and estrogen production, a major risk factor for breast cancer in obese postmenopausal women. TNF-α, a proinflammatory cytokine, is a key driver of aromatase promoter I.4-mediated expression in adipose tissue. In this study, we have shown that IL-10, an anti-inflammatory cytokine, suppressed both TNF-α-stimulated human aromatase reporter-luciferase (hARO-Luc) expression in mouse bone marrow mesenchymal stromal cells and aromatase gene expression in human breast adipose stromal cells (ASCs). IL-10 blocked TNF-α-stimulated ERK1/2 activation in ASCs, suggesting an inhibitory effect through the MAPK signaling pathway. The links among obesity, IL-10, and aromatase were confirmed in ovariectomized (OVX) hARO-Luc mice, where increased adiposity was associated with upregulation of aromatase reporter activity and reduced IL-10 level in the mammary fat pad. OVX mice also exhibited changes in gut microbiota, similar to that in obese women, indicating altered immune function. In summary, our results suggest that increased adiposity, induced by the lack of ovarian hormones, results in enhanced expression of aromatase in mammary adipose tissue, mediated by reduction in local IL-10. These findings may bring new insights into the mechanisms involved in the development of postmenopausal breast cancer, as well as novel approaches for prevention.-Martínez-Chacón, G., Brown, K. A., Docanto, M. M., Kumar, H., Salminen, S., Saarinen, N., Mäkelä, S. IL-10 suppresses TNF-α-induced expression of human aromatase gene in mammary adipose tissue.


Assuntos
Tecido Adiposo/enzimologia , Aromatase/biossíntese , Mama/enzimologia , Regulação Enzimológica da Expressão Gênica , Interleucina-10/metabolismo , Sistema de Sinalização das MAP Quinases , Fator de Necrose Tumoral alfa/metabolismo , Animais , Feminino , Humanos , Glândulas Mamárias Animais/enzimologia , Camundongos , Camundongos Transgênicos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo
5.
Nat Prod Rep ; 32(8): 1249-66, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26030402

RESUMO

It is widely accepted that drug discovery often requires a systems-level polypharmacology approach to tackle problems such as lack of efficacy and emerging resistance of single-targeted compounds. Network pharmacology approaches are increasingly being developed and applied to find new therapeutic opportunities and to re-purpose approved drugs. However, these recent advances have been relatively slow to be translated into the field of natural products. Here, we argue that a network pharmacology approach would enable an effective mapping of the yet unexplored target space of natural products, hence providing a systematic means to extend the druggable space of proteins implicated in various complex diseases. We give an overview of the key network pharmacology concepts and recent experimental-computational approaches that have been successfully applied to natural product research, including unbiased elucidation of mechanisms of action as well as systematic prediction of effective therapeutic combinations. We focus specifically on anticancer applications that use in vivo and in vitro functional phenotypic measurements, such as genome-wide transcriptomic response profiles, which enable a global modelling of the multi-target activity at the level of the biological pathways and interaction networks. We also provide representative examples of other disease applications, databases and tools as well as existing and emerging resources, which may prove useful for future natural product research. Finally, we offer our personal view of the current limitations, prospective developments and open questions in this exciting field.


Assuntos
Produtos Biológicos , Produtos Biológicos/farmacologia , Biologia Computacional , Descoberta de Drogas , Humanos , Estrutura Molecular
6.
Am J Pathol ; 184(8): 2163-73, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24949550

RESUMO

Androgens are key factors involved in the development and progression of prostate cancer (PCa), and PCa growth can be suppressed by androgen deprivation therapy. In a considerable proportion of men receiving androgen deprivation therapy, however, PCa progresses to castration-resistant PCa (CRPC), making the development of efficient therapies challenging. We used an orthotopic VCaP human PCa xenograft model to study cellular and molecular changes in tumors after androgen deprivation therapy (castration). Tumor growth was monitored through weekly serum prostate-specific antigen measurements, and mice with recurrent tumors after castration were randomized to treatment groups. Serum prostate-specific antigen concentrations showed significant correlation with tumor volume. Castration-resistant tumors retained concentrations of intratumoral androgen (androstenedione, testosterone, and 5α-dihydrotestosterone) at levels similar to tumors growing in intact hosts. Accordingly, castration induced up-regulation of enzymes involved in androgen synthesis (CYP17A1, AKR1C3, and HSD17B6), as well as expression of full-length androgen receptor (AR) and AR splice variants (AR-V1 and AR-V7). Furthermore, AR target gene expression was maintained in castration-resistant xenografts. The AR antagonists enzalutamide (MDV3100) and ARN-509 suppressed PSA production of castration-resistant tumors, confirming the androgen dependency of these tumors. Taken together, the findings demonstrate that our VCaP xenograft model exhibits the key characteristics of clinical CRPC and thus provides a valuable tool for identifying druggable targets and for testing therapeutic strategies targeting AR signaling in CRPC.


Assuntos
Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias da Próstata , Androgênios/biossíntese , Animais , Antineoplásicos Hormonais/farmacologia , Castração , Xenoenxertos , Humanos , Immunoblotting , Masculino , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores Androgênicos/biossíntese , Regulação para Cima
7.
FASEB J ; 27(4): 1342-50, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23239821

RESUMO

The human CYP19A1 gene is expressed in various tissues by the use of tissue-specific promoters, whereas the rodent cyp19a1 gene is expressed mainly in the gonads and brain. We generated a transgenic mouse model containing a >100-kb 5' region of human CYP19A1 gene connected to a luciferase reporter gene. The luciferase activity in mouse tissues mimicked the CYP19A1 gene expression pattern in humans. Interestingly, the reporter gene activity was 16 and 160 times higher in the urinary bladder and seminal vesicles, respectively, as compared with the activity in the testis. Accordingly, CYP19A1 gene and P450arom protein expression was detected in those human tissues. Moreover, the data revealed that the expression of CYP19A1 gene is driven by promoters PII, I.4, and I.3 in the seminal vesicles, and by promoters PII and I.4 in the urinary bladder. Furthermore, the reporter gene expression in the seminal vesicles was androgen dependent: Castration decreased the expression ∼20 times, and testosterone treatment restored it to the level of an intact mouse. This reporter mouse model facilitates studies of tissue-specific regulation of the human CYP19A1 gene, and our data provide evidence for seminal vesicles as important sites for estrogen production in males.


Assuntos
Androgênios/metabolismo , Aromatase/metabolismo , Glândulas Seminais/metabolismo , Bexiga Urinária/metabolismo , Androgênios/genética , Animais , Aromatase/genética , Regulação Enzimológica da Expressão Gênica/genética , Genes Reporter/genética , Humanos , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Sequências Reguladoras de Ácido Nucleico/genética , Testículo/metabolismo
8.
Oncogenesis ; 12(1): 42, 2023 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-37573448

RESUMO

Lethal prostate cancer (PCa) is characterized by the presence of metastases and development of resistance to therapies. Metastases form in a multi-step process enabled by dynamic cytoskeleton remodeling. An actin cytoskeleton regulating gene, CALD1, encodes a protein caldesmon (CaD). Its isoform, low-molecular-weight CaD (l-CaD), operates in non-muscle cells, supporting the function of filaments involved in force production and mechanosensing. Several factors, including glucocorticoid receptor (GR), have been identified as regulators of l-CaD in different cell types, but the regulation of l-CaD in PCa has not been defined. PCa develops resistance in response to therapeutic inhibition of androgen signaling by multiple strategies. Known strategies include androgen receptor (AR) alterations, modified steroid synthesis, and bypassing AR signaling, for example, by GR upregulation. Here, we report that in vitro downregulation of l-CaD promotes epithelial phenotype and reduces spheroid growth in 3D, which is reflected in vivo in reduced formation of metastases in zebrafish PCa xenografts. In accordance, CALD1 mRNA expression correlates with epithelial-to-mesenchymal transition (EMT) transcripts in PCa patients. We also show that CALD1 is highly co-expressed with GR in multiple PCa data sets, and GR activation upregulates l-CaD in vitro. Moreover, GR upregulation associates with increased l-CaD expression after the development of resistance to antiandrogen therapy in PCa xenograft mouse models. In summary, GR-regulated l-CaD plays a role in forming PCa metastases, being clinically relevant when antiandrogen resistance is attained by the means of bypassing AR signaling by GR upregulation.

9.
Am J Pathol ; 179(3): 1148-56, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21763263

RESUMO

In this study, we investigated the effects of ectopic estrogen receptor (ER)ß1 expression in breast cancer cell lines and nude mice xenografts and observed that ERß1 expression suppresses tumor growth and represses FOXM1 mRNA and protein expression in ERα-positive but not ERα-negative breast cancer cells. Furthermore, a significant inverse correlation exists between ERß1 and FOXM1 expression at both protein and mRNA transcript levels in ERα-positive breast cancer patient samples. Ectopic ERß1 expression resulted in decreased FOXM1 protein and mRNA expression only in ERα-positive but not ERα-negative breast carcinoma cell lines, suggesting that ERß1 represses ERα-dependent FOXM1 transcription. Reporter gene assays showed that ERß1 represses FOXM1 transcription through an estrogen-response element located within the proximal promoter region that is also targeted by ERα. The direct binding of ERß1 to the FOXM1 promoter was confirmed by chromatin immunoprecipitation analysis, which also showed that ectopic expression of ERß1 displaces ERα from the endogenous FOXM1 promoter. Forced expression of ERß1 promoted growth suppression in MCF-7 cells, but the anti-proliferative effects of ERß1 could be overridden by overexpression of FOXM1, indicating that FOXM1 is an important downstream target of ERß1 signaling. Together, these findings define a key anti-proliferative role for ERß1 in breast cancer development through negatively regulating FOXM1 expression.


Assuntos
Neoplasias da Mama/patologia , Proliferação de Células , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Proteína Forkhead Box M1 , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Transplante de Neoplasias , RNA Mensageiro/metabolismo , Transcrição Gênica , Transplante Heterólogo
10.
Behav Genet ; 42(4): 509-27, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22426781

RESUMO

Inspired by the localization, on 15q21.2 of the CYP19A1 gene in the linkage region of speech and language disorders, and a rare translocation in a dyslexic individual that was brought to our attention, we conducted a series of studies on the properties of CYP19A1 as a candidate gene for dyslexia and related conditions. The aromatase enzyme is a member of the cytochrome P450 super family, and it serves several key functions: it catalyzes the conversion of androgens into estrogens; during early mammalian development it controls the differentiation of specific brain areas (e.g. local estrogen synthesis in the hippocampus regulates synaptic plasticity and axonal growth); it is involved in sexual differentiation of the brain; and in songbirds and teleost fishes, it regulates vocalization. Our results suggest that variations in CYP19A1 are associated with dyslexia as a categorical trait and with quantitative measures of language and speech, such as reading, vocabulary, phonological processing and oral motor skills. Variations near the vicinity of its brain promoter region altered transcription factor binding, suggesting a regulatory role in CYP19A1 expression. CYP19A1 expression in human brain correlated with the expression of dyslexia susceptibility genes such as DYX1C1 and ROBO1. Aromatase-deficient mice displayed increased cortical neuronal density and occasional cortical heterotopias, also observed in Robo1-/- mice and human dyslexic brains, respectively. An aromatase inhibitor reduced dendritic growth in cultured rat neurons. From this broad set of evidence, we propose CYP19A1 as a candidate gene for human cognitive functions implicated in reading, speech and language.


Assuntos
Aromatase/genética , Encéfalo/crescimento & desenvolvimento , Dislexia/genética , Transtornos da Linguagem/genética , RNA Mensageiro/análise , Distúrbios da Fala/genética , Animais , Aromatase/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Coortes , Proteínas do Citoesqueleto , Dislexia/metabolismo , Feminino , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Transtornos da Linguagem/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Locos de Características Quantitativas , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Distúrbios da Fala/metabolismo , Translocação Genética , Proteínas Roundabout
11.
iScience ; 25(5): 104287, 2022 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-35573198

RESUMO

Antiandrogen treatment resistance is a major clinical concern in castration-resistant prostate cancer (CRPC) treatment. Using xenografts of VCaP cells we showed that growth of antiandrogen resistant CRPC tumors were characterized by a higher intratumor dihydrotestosterone (DHT) concentration than that of treatment responsive tumors. Furthermore, the slow tumor growth after adrenalectomy was associated with a low intratumor DHT concentration. Reactivation of androgen signaling in enzalutamide-resistant tumors was further shown by the expression of several androgen-dependent genes. The data indicate that intratumor DHT concentration and expression of several androgen-dependent genes in CRPC lesions is an indication of enzalutamide treatment resistance and an indication of the need for further androgen blockade. The presence of an androgen synthesis, independent of CYP17A1 activity, has been shown to exist in prostate cancer cells, and thus, novel androgen synthesis inhibitors are needed for the treatment of enzalutamide-resistant CRPC tumors that do not respond to abiraterone.

12.
Am J Pathol ; 176(3): 1443-51, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20093485

RESUMO

Local estrogen production plays a key role in proliferative endometrial disorders, such as endometrial hyperplasia and cancer. Hydroxysteroid (17beta) dehydrogenase 1 (HSD17B1) is an enzyme that catalyzes with high efficiency the conversion of weakly active estrone into highly potent estradiol. Here we report that female transgenic mice expressing human HSD17B1 invariably develop endometrial hyperplasia in adulthood. These mice also fail to ovulate and have enhanced peripheral conversion of estrone into estradiol in a variety of target tissues, including the uterus. As in humans, endometrial hyperplasia in HSD17B1 transgenic female mice was reversible on ovulation induction, which triggers a rise in circulating progesterone levels, and in response to exogenous progestins. Strikingly, a treatment with an HSD17B1 inhibitor failed to restore ovulation yet completely reversed the hyperplastic morphology of epithelial cells in the glandular compartment, although less so in the luminal epithelium. The data indicate that human HSD17B1 expression enhances endometrial estrogen production, and consequently, estrogen-dependent proliferation. Therefore, HSD17B1 is a promising new therapeutic target in the management of estrogen-dependent endometrial diseases.


Assuntos
17-Hidroxiesteroide Desidrogenases/antagonistas & inibidores , Hiperplasia Endometrial/tratamento farmacológico , Hiperplasia Endometrial/patologia , Inibidores Enzimáticos/uso terapêutico , Animais , Modelos Animais de Doenças , Hiperplasia Endometrial/enzimologia , Inibidores Enzimáticos/farmacologia , Estradiol/metabolismo , Estrogênios , Feminino , Humanos , Camundongos , Camundongos Transgênicos , Tamanho do Órgão/efeitos dos fármacos , Progestinas/uso terapêutico , Útero/efeitos dos fármacos , Útero/enzimologia , Útero/crescimento & desenvolvimento , Útero/patologia
13.
J Nutr ; 141(9): 1583-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21753063

RESUMO

Enterolactone (EL) is an enterolignan produced by gut microbiota from dietary plant lignans. Epidemiological and experimental studies suggest that EL and plant lignans may reduce the risk of breast and prostate cancer as well as cardiovascular disease. These effects are thought to at least in part involve modulation of estrogen receptor activity. Surprisingly little is known about the in vivo estrogenicity of EL. In the present study, we investigated the target tissues of EL, the genes affected by EL treatment, and the response kinetics. Following a single dose of EL, luciferase was significantly induced in reproductive and nonreproductive tissues of male and female 3xERE-luciferase mice, indicating estrogen-like activity. Microarray analysis revealed that EL regulated the expression of only 1% of 17ß-estradiol target genes in the uterus. The majority of these genes were traditional estrogen target genes, but also members of the circadian signaling pathway were affected. Kinetic analyses showed that EL undergoes rapid phase II metabolism and is efficiently excreted. In vivo imaging demonstrated that the estrogen response followed similar, fast kinetics. We conclude that EL activates estrogen signaling in both male and female mice and that the transient responses may be due to the fast metabolism of the compound. Lastly, EL may represent a link among diet, gut microbiota, and circadian signaling.


Assuntos
4-Butirolactona/análogos & derivados , Proteínas CLOCK/metabolismo , Relógios Circadianos/genética , Estrogênios/metabolismo , Lignanas/farmacologia , Fitoestrógenos/farmacologia , Transdução de Sinais/efeitos dos fármacos , 4-Butirolactona/sangue , 4-Butirolactona/farmacologia , Animais , Proteínas CLOCK/genética , Relógios Circadianos/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Lignanas/sangue , Fígado/metabolismo , Luciferases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Orquiectomia , Ovariectomia , Análise Serial de Proteínas , Distribuição Aleatória , Útero/metabolismo
14.
Reprod Health ; 7: 26, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20932282

RESUMO

BACKGROUND: Mother's diet during pregnancy is important, since plant lignans and their metabolites, converted by the intestinal microflora to enterolignans, are proposed to possess multiple health benefits. Aim of our study was to investigate whether a dietary intervention affects lignan concentrations in the serum of pregnant women. METHODS: A controlled dietary intervention trial including 105 first-time pregnant women was conducted in three intervention and three control maternity health clinics. The intervention included individual counseling on diet and on physical activity, while the controls received conventional care. Blood samples were collected on gestation weeks 8-9 (baseline) and 36-37 (end of intervention). The serum levels of the plant lignans 7-hydroxymatairesinol, secoisolariciresinol, matairesinol, lariciresinol, cyclolariciresinol, and pinoresinol, and of the enterolignans 7-hydroxyenterolactone, enterodiol, and enterolactone, were measured using a validated method. RESULTS: The baseline levels of enterolactone, enterodiol and the sum of lignans were higher in the control group, whereas at the end of the trial their levels were higher in the intervention group. The adjusted mean differences between the baseline and end of the intervention for enterolactone and the total lignan intake were 1.6 ng/ml (p = 0.018, 95% CI 1.1-2.3) and 1.4 ng/mg (p = 0.08, 95% CI 1.0-1.9) higher in the intervention group than in the controls. Further adjustment for dietary components did not change these associations. CONCLUSION: The dietary intervention was successful in increasing the intake of lignan-rich food products, the fiber consumption and consequently the plasma levels of lignans in pregnant women. TRIAL REGISTRATION: ISRCTN21512277, http://www.isrctn.org.

15.
EXS ; 99: 289-323, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19157066

RESUMO

Many toxic compounds exert their harmful effects by activating of certain receptors, which in turn leads to dysregulation of transcription. Some of these receptors are so called xenosensors. They are activated by external chemicals and evoke a cascade of events that lead to the elimination of the chemical from the system. Other receptors that are modulated by toxic substances are hormone receptors, particularly the ones of the nuclear receptor family. Some environmental chemicals resemble endogenous hormones and can falsely activate these receptors, leading to undesired activity in the cell. Furthermore, excessive activation of the xenosensors can lead to disturbances of the integrity of the system as well. In this chapter, the concepts of receptor-mediated toxicity and hormone disruption are introduced. We start by describing environmental chemicals that can bind to xenosensors and nuclear hormone receptors. We then describe the receptors most commonly targeted by environmental chemicals. Finally, the mechanisms by which receptor-mediated events can disrupt the system are depicted.


Assuntos
Disruptores Endócrinos/toxicidade , Sistema Endócrino/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Sistema Endócrino/metabolismo , Sistema Endócrino/fisiologia , Humanos , Modelos Biológicos , Receptores Citoplasmáticos e Nucleares/fisiologia , Transdução de Sinais/efeitos dos fármacos , Xenobióticos/toxicidade
16.
Mol Cell Endocrinol ; 301(1-2): 163-8, 2009 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-19061935

RESUMO

When present in excess amounts during fetal life, androgens can impair female development by inducing masculinization. On way to modify fetal steroid concentration is by altering the expression of hydroxysteroid (17beta) dehydrogenases (HSD17Bs). Human HSD17B1 converts weak estrogen estrone to estradiol, and with lower catalytic efficiency, weak androgen androstenedione to testosterone. We have recently shown that over-expression of human HSD17B1 in transgenic mice results in masculinized phenotype in female mice. In the present study, we further show that in addition to the Müllerian ducts, HSD17B1TG females have internal structures resembling Wolffian ducts, and enlarged Skene paraurethral gland, also called the female prostate. HSD17B1 expression has been found in fetal human ovary, thus, it is possible that HSD17B1 contributes to maintain the normal steroid hormone concentration during development. Thereby, abnormal increase in the fetal expression of HSD17B1 could contribute to the development of hormonal imbalances, and so result in female masculinization.


Assuntos
Estradiol Desidrogenases/metabolismo , Virilismo/enzimologia , Animais , Feminino , Humanos , Hipertrofia , Camundongos , Camundongos Transgênicos , Ductos Mesonéfricos/patologia
17.
Int J Cancer ; 123(5): 1196-204, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18528864

RESUMO

Lariciresinol is a dietary lignan that accounts for a significant portion of the total phytoestrogen intake from Western foods. Recent epidemiological studies suggest that high dietary intake of lignans and lariciresinol is associated with reduced breast cancer risk. However, no causal relationship between lariciresinol intake and breast cancer development has been established. In this study, we investigated for the first time the effects and possible mechanisms of action of lariciresinol on hormone responsive mammary cancer in vivo in dimethylbenz[a]anthracene induced mammary cancer in rats, and in human MCF-7 breast cancer xenografts in athymic mice. For tumor bearing rats, lariciresinol (3 or 15 mg/kg of body weight) or vehicle was administered p.o. daily for 9 weeks. For E2-maintained ovariectomized athymic mice bearing orthotopic MCF-7 tumors, control diet (AIN-93G) or lariciresinol containing diet (AIN-93G supplemented with 20 or 100 mg of lariciresinol/kg of diet) was administered for 5 weeks. In both models, lariciresinol administration inhibited the tumor growth and tumor angiogenesis. In MCF-7 cells, enterolactone significantly inhibited the E2-stimulated VEGF secretion. Moreover, in MCF-7 xenografts, lariciresinol administration enhanced tumor cell apoptosis and increased estrogen receptor beta expression. Lariciresinol and its further metabolites secoisolariciresinol, enterodiol and enterolactone were found in serum of both rats and athymic mice confirming a similar lignan metabolism pattern as in humans. These findings indicate conceivable importance of dietary lignan lariciresinol in inhibition of breast cancer development.


Assuntos
Antineoplásicos/farmacologia , Suplementos Nutricionais , Furanos/farmacologia , Lignanas/sangue , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/patologia , 4-Butirolactona/análogos & derivados , 4-Butirolactona/sangue , 9,10-Dimetil-1,2-benzantraceno , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Butileno Glicóis/sangue , Carcinógenos , Proliferação de Células/efeitos dos fármacos , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Feminino , Furanos/sangue , Furanos/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Lignanas/farmacologia , Lignanas/uso terapêutico , Neoplasias Mamárias Experimentais/sangue , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/induzido quimicamente , Camundongos , Camundongos Nus , Ovariectomia , Fitoestrógenos/sangue , Ratos , Receptores de Progesterona/metabolismo , Transplante Heterólogo
18.
Endocr Relat Cancer ; 25(9): 807-819, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29773553

RESUMO

Intratumoral androgen biosynthesis is one of the mechanisms involved in the progression of prostate cancer, and an important target for novel prostate cancer therapies. Using gas chromatography-tandem mass spectrometry and genome-wide RNA sequencing, we have analyzed androgen concentrations and androgen-regulated gene expression in cancerous and morphologically benign prostate tissue specimens and serum samples obtained from 48 primary prostate cancer patients. Intratumoral dihydrotestosterone (DHT) concentrations were significantly higher in the cancerous tissues compared to benign prostate (P < 0.001). The tissue/serum ratios of androgens were highly variable between the patients, indicating individual patterns of androgen metabolism and/or uptake of androgens within the prostate tissue. An unsupervised hierarchical clustering analysis of intratissue androgen concentrations indicated that transmembrane protease, serine 2/ETS-related gene (TMPRSS2-ERG)-positive patients have different androgen profiles compared to TMPRSS2-ERG-negative patients. TMPRSS2-ERG gene fusion status was also associated with an enhanced androgen-regulated gene expression, along with altered intratumoral androgen metabolism, demonstrated by reduced testosterone concentrations and increased DHT/testosterone ratios in TMPRSS2-ERG-positive tumors. TMPRSS2-ERG-positive and -negative prostate cancer specimens have distinct intratumoral androgen profiles, possibly due to activation of testosterone-independent DHT biosynthesis via the alternative pathway in TMPRSS2-ERG-positive tumors. Thus, patients with TMPRSS2-ERG-positive prostate cancer may benefit from novel inhibitors targeting the alternative DHT biosynthesis.


Assuntos
Androgênios/metabolismo , Di-Hidrotestosterona/metabolismo , Proteínas de Fusão Oncogênica/genética , Neoplasias da Próstata , Serina Endopeptidases/genética , Testosterona/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Próstata/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Regulador Transcricional ERG/genética
19.
Endocrinology ; 148(10): 4875-86, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17628008

RESUMO

Numerous dietary compounds can modify gene expression by binding to the members of the nuclear receptor superfamily of transcription factors. For example, dietary polyphenols, such as soy isoflavones genistein and daidzein, modulate the activity of the estrogen receptors (ERs)-alpha and ERbeta. An additional class of dietary polyphenols that modulate cellular signaling pathways are lignans, compounds that are common constituents of Western diets. In this study, we show that a metabolite of dietary lignans, enterolactone, at physiological concentrations, activates ER-mediated transcription in vitro with preference for ERalpha. The effects of enterolactone are mediated by the ER ligand binding domain and are susceptible to antiestrogen treatment. Furthermore, the affinity of enterolactone toward ERalpha, measured by a novel ligand binding assay, is augmented in cell culture conditions. Moreover, our results demonstrate for the first time that enterolactone has estrogenic activity in vivo. In transgenic estrogen-sensitive reporter mice, enterolactone induces tissue-specific estrogen-responsive reporter gene expression as well as promotes uterine stromal edema and expression of estrogen-responsive endogenous genes (CyclinD1 and Ki67). Taken together, our data show that enterolactone is a selective ER agonist inducing ER-mediated transcription both in vitro in different cell lines and in vivo in the mouse uterus.


Assuntos
4-Butirolactona/análogos & derivados , Dieta , Lignanas/metabolismo , Receptores de Estrogênio/efeitos dos fármacos , Receptores de Estrogênio/metabolismo , 4-Butirolactona/biossíntese , 4-Butirolactona/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/metabolismo , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Expressão Gênica/efeitos dos fármacos , Genes Reporter , Humanos , Ligantes , Lignanas/biossíntese , Lignanas/farmacologia , Camundongos , Camundongos Transgênicos , Estrutura Terciária de Proteína/fisiologia , Receptores de Estrogênio/agonistas , Distribuição Tecidual , Ativação Transcricional/efeitos dos fármacos
20.
Mol Nutr Food Res ; 51(7): 857-66, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17576639

RESUMO

Lignans are a large group of fiber-associated phenolic compounds widely distributed in edible plants. Some of the ingested plant lignans are converted by intestinal microbiota to enterolignans, enterodiol (END) and enterolactone (ENL), the latter of which has been thought to be the major biologically active lignan, and suggested to be associated with low risk of breast cancer. In line with this, administration of plant lignans which are further metabolized to ENL, or ENL as such, have been shown to inhibit or delay the growth of experimental mammary cancer. The mechanism of anticarcinogenic action of ENL is not yet fully understood, but there is intriguing evidence for ENL as a modulator of estrogen signaling. These findings have generated interest in the use of lignans as components of breast cancer risk reducing functional foods. Identification of target groups, who would benefit most, is of pivotal importance. Therefore, further identification and validation of relevant biomarkers, which can be used as indicators of lignan or ENL action and breast cancer risk reduction at different stages of the disease, are of importance.


Assuntos
Anticarcinógenos/administração & dosagem , Neoplasias da Mama/prevenção & controle , Dieta , Lignanas/administração & dosagem , 4-Butirolactona/análogos & derivados , 4-Butirolactona/metabolismo , 4-Butirolactona/farmacologia , Animais , Bactérias/metabolismo , Neoplasias da Mama/patologia , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Intestinos/microbiologia , Lignanas/metabolismo , Lignanas/farmacologia , Neoplasias Mamárias Animais/prevenção & controle , Plantas Comestíveis/química , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa