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

Medicinas Complementares
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Int J Clin Pract ; 2022: 1263544, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36531558

RESUMO

Objective: The purpose of this study was to evaluate the impact of isoflavone supplementation compared with placebo on endometrial histology and serum estradiol levels in premenopausal women with nonatypical endometrial hyperplasia. Materials and Methods: The present double-blindplacebo-controlled clinical trial was conducted on 100 women with nonatypical endometrial hyperplasia in the age range of 30 to 45 years. Participants were randomly assigned to receive 50 mg of isoflavone (n = 50) or placebos (n = 50) daily for three months. Both groups received the standard treatment of nonatypical endometrial hyperplasia. Endometrial biopsy and blood samples were taken at the baseline and three months after the intervention. The incidence of drug side effects was assessed as well. Results: After three months, 88.4% of isoflavone-administered subjects had a significant histological improvement compared to 68.9% subjects in the placebo group (P=0.02). There were no significant differences between the two groups in the changes of serum estradiol levels and the incidence of drug side effects. Conclusion: The findings of the present study demonstrated that the coadministration of 50 mg of isoflavones and medroxyprogesterone acetate increases the treatment efficacy in women with nonatypical endometrial hyperplasia. Clinical Trial Registration. This trial was registered on the Iranian website for clinical trial registration (https://www.irct.ir/trial/53553).


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Hiperplasia Endometrial , Isoflavonas , Feminino , Humanos , Adulto , Pessoa de Meia-Idade , Hiperplasia Endometrial/tratamento farmacológico , Hiperplasia Endometrial/induzido quimicamente , Hiperplasia Endometrial/epidemiologia , Isoflavonas/efeitos adversos , Medroxiprogesterona , Irã (Geográfico) , Método Duplo-Cego , Estradiol/efeitos adversos , Suplementos Nutricionais
2.
Gen Comp Endocrinol ; 266: 126-134, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29777688

RESUMO

Although neonatal exposure to estrogen or estrogenic compounds results in irreversible changes in the brain function and reproductive abnormalities during adulthood but the underlying mechanisms are still largely unknown. The present study has attempted to compare the protein profiles of sexually dimorphic brain regions of adult female rats which were exposed to estradiol- 17ß during neonatal period. The total proteins extracted from pre-optic area (POA), hypothalamus, hippocampus and pituitary of control and neonatally E2 treated female rats was subjected to 2D-SDS-PAGE and differentially expressed proteins were identified by MALDI TOF/TOF-MS. Our results revealed that a total of 21 protein spots which were identified as differentially expressed in all the four regions analyzed; the differential expression was further validated by RT-PCR and western blotting. The differentially expressed proteins such as 14-3-3 zeta/delta (POA), LMNA (hippocampus), Axin2 (hypothalamus), Syntaxin-7 (hippocampus), prolactin and somatotropin (pituitary) which have very important functions in the process of neuronal differentiation, migration, axon outgrowth, formation of dendritic spine density and synaptic plasticity and memory have not been previously reported in association with neonatal estrogen exposure. The affected brain functions are very important for the establishment of sex specific brain morphology and behavior. Our results suggest that the differentially expressed proteins may play an important role in irreversible changes in the brain function as well as reproductive abnormalities observed in the female rats during adulthood.


Assuntos
Envelhecimento/metabolismo , Estradiol/efeitos adversos , Hipocampo/metabolismo , Hipotálamo/metabolismo , Hipófise/metabolismo , Área Pré-Óptica/metabolismo , Proteômica/métodos , Animais , Animais Recém-Nascidos , Regulação para Baixo , Feminino , Ontologia Genética , Redes Reguladoras de Genes , Espectrometria de Massas , Proteoma/metabolismo , Ratos , Reprodutibilidade dos Testes , Regulação para Cima
3.
Endocrinology ; 159(1): 465-476, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29155986

RESUMO

An adverse intrauterine environment may induce adult disease in offspring, but the mechanisms are not well understood. It is reported that fresh embryo transfer (ET) in assisted reproductive technology leads to high maternal estradiol (E2), and prenatal high E2 exposure increases the risk of organ disorders in later life. We found that male newborns and children of fresh ET showed elevated fasting insulin and homeostasis model of assessment for insulin resistance index (HOMA-IR) scores. Male mice with high prenatal estradiol exposure (HE) grew heavier than control mice and developed insulin resistance; they also showed increased food intake, with increased orexigenic hypothalamic neuropeptide Y (NPY) expression. The hypothalamic insulin receptor (INSR) was decreased in male HE mice, associated with elevated promoter methylation. Chronic food restriction (FR) in HE mice reversed insulin resistance and rescued hypothalamic INSR expression by correcting the elevated Insr promoter methylation. Our findings suggest that prenatal exposure to high E2 may induce sex-specific metabolic disorders in later life through epigenetic programming of hypothalamic Insr promoter, and dietary intervention may reverse insulin resistance by remodeling its methylation pattern.


Assuntos
Estradiol/efeitos adversos , Fármacos para a Fertilidade Feminina/efeitos adversos , Hiperinsulinismo/induzido quimicamente , Hipotálamo/efeitos dos fármacos , Resistência à Insulina , Neurônios/efeitos dos fármacos , Efeitos Tardios da Exposição Pré-Natal , Animais , Criança , Pré-Escolar , Transferência Embrionária/efeitos adversos , Ingestão de Energia/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Recém-Nascido , Masculino , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/metabolismo , Neuropeptídeo Y/agonistas , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , Gravidez , Distribuição Aleatória , Receptor de Insulina/antagonistas & inibidores , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Aumento de Peso/efeitos dos fármacos
4.
J Biochem Mol Toxicol ; 32(1)2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28960787

RESUMO

The objective of the present study was to characterize the role of novel resveratrol (Res) analogs: 4-(E)-{(4-hydroxyphenylimino)-methylbenzene, 1, 2-diol} (HPIMBD) and 4-(E)-{(p-tolylimino)-methylbenzene-1,2-diol} (TIMBD) as potent antioxidants against breast cancer. Non-neoplastic breast epithelial cell lines MCF-10A and MCF-10F were treated with 17ß-estradiol (E2), Res, HPIMBD, and TIMBD for up to 72 h. mRNA and protein levels of antioxidant genes, superoxide dismutase 3 (SOD3) and N-quinoneoxidoreductase-1 (NQO1) and transcription factors, nuclear factor erythroid 2-related factor (Nrf) 1, 2 and 3 were quantified after the above treatments. Generation of reactive oxygen species (ROS) was measured by CM-H2-DCFDA and oxidative-DNA damage was determined by measuring 8-hydroxy-2-deoxyguanosine (8-OHdG). HPIMBD and TIMBD scavenged cellular ROS production, attenuated oxidative DNA damage, increased mRNA and protein expression levels of SOD3 and NQO1 and activated Nrf signaling pathway. Our studies demonstrate that HPIMBD and TIMBD have the potential as novel antioxidants to prevent development of breast cancer.


Assuntos
Anticarcinógenos/metabolismo , Antioxidantes/metabolismo , Neoplasias da Mama/prevenção & controle , Mama/metabolismo , Catecóis/metabolismo , Bases de Schiff/metabolismo , Estilbenos/metabolismo , 8-Hidroxi-2'-Desoxiguanosina , Anticarcinógenos/efeitos adversos , Antioxidantes/efeitos adversos , Mama/citologia , Mama/patologia , Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Catecóis/efeitos adversos , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , Dano ao DNA , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Suplementos Nutricionais/efeitos adversos , Indução Enzimática , Estradiol/efeitos adversos , Feminino , Humanos , NAD(P)H Desidrogenase (Quinona)/química , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Resveratrol , Bases de Schiff/efeitos adversos , Transdução de Sinais , Estilbenos/efeitos adversos , Superóxido Dismutase/química , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
5.
Afr J Tradit Complement Altern Med ; 14(4): 247-252, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28638887

RESUMO

BACKGROUND: Mammary hyperplasia is one of the most common benign breast disorders. Although traditional Chinese medicine has a vast experience in the treatment of mammary hyperplasia, it is not accepted widely due to its unclear mechanism. METHODS AND MATERIALS: To address the mechanism, we developed a mouse model of mammary hyperplasia. We gave mice estradiol valerate tablets and progesterone capsules sequentially for one month by intragastric administration. RESULTS: Mice treated by this method had a series of pathological changes which are similar to those detected in women with mammary hyperplasia, including ectopic level of estradiol and progesterone in serum, hyperplasia of mammary glands and increased expression of ERα and PR. CONCLUSION: This model will facilitate the mechanical study of traditional medicine on mammary hyperplasia.


Assuntos
Doenças Mamárias/patologia , Modelos Animais de Doenças , Estradiol/análogos & derivados , Hiperplasia/patologia , Progesterona/efeitos adversos , Administração Oral , Animais , Doenças Mamárias/induzido quimicamente , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Hiperplasia/induzido quimicamente , Camundongos , Camundongos Endogâmicos C57BL , Progesterona/administração & dosagem , Receptores de Progesterona/genética , Receptores de Progesterona/metabolismo
6.
Int J Mol Sci ; 18(2)2017 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-28212313

RESUMO

Scientific evidence suggests that many herbs and spices have medicinal properties that alleviate symptoms or prevent disease. In this study, we examined the chemopreventive effects of the Apiaceae spices, anise, caraway, and celery seeds against 17ß-estrogen (E2)-mediated mammary tumorigenesis in an ACI (August-Copenhagen Irish) rat model. Female ACI rats were given either control diet (AIN 93M) or diet supplemented with 7.5% (w/w) of anise, caraway, or celery seed powder. Two weeks later, one half of the animals in each group received subcutaneous silastic implants of E2. Diet intake and body weight were recorded weekly, and animals were euthanized after 3 and 12 weeks. E2-treatment showed significantly (2.1- and 3.4-fold) enhanced growth of pituitary gland at 3 and 12 weeks, respectively. All test spices significantly offset the pituitary growth by 12 weeks, except celery which was effective as early as three weeks. Immunohistochemical analysis for proliferative cell nuclear antigen (PCNA) in mammary tissues showed significant reduction in E2-mediated mammary cell proliferation. Test spices reduced the circulating levels of both E2 and prolactin at three weeks. This protection was more pronounced at 12 weeks, with celery eliciting the highest effect. RT-PCR and western blot analysis were performed to determine the potential molecular targets of the spices. Anise and caraway diets significantly offset estrogen-mediated overexpression of both cyclin D1 and estrogen receptor α (ERα). The effect of anise was modest. Likewise, expression of CYP1B1 and CYP1A1 was inhibited by all test spices. Based on short-term molecular markers, caraway was selected over other spices based on its enhanced effect on estrogen-associated pathway. Therefore, a tumor-end point study in ACI rats was conducted with dietary caraway. Tumor palpation from 12 weeks onwards revealed tumor latency of 29 days in caraway-treated animals compared with first tumor appearance at 92 days in control group. At the end of the study (25 weeks), the tumor incidence was 96% in the control group compared with only 70% in the caraway group. A significant reduction in tumor volume (661 ± 123 vs. 313 ± 81 mm³) and tumor multiplicity (4.2 ± 0.4 vs. 2.5 ± 0.5 tumors/animal) was also observed in the caraway group compared with the control group. Together, our data show dietary caraway can significantly delay and prevent the hormonal mammary tumorigenesis by modulating different cellular and molecular targets.


Assuntos
Apiaceae/química , Transformação Celular Neoplásica/efeitos dos fármacos , Quimioprevenção , Suplementos Nutricionais , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/prevenção & controle , Especiarias , Animais , Biomarcadores , Peso Corporal , Proliferação de Células/efeitos dos fármacos , Estradiol/efeitos adversos , Estrogênios/sangue , Feminino , Neoplasias Mamárias Experimentais/sangue , Prolactina/sangue , Ratos , Carga Tumoral
7.
Cochrane Database Syst Rev ; 1: CD011093, 2017 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-28043088

RESUMO

BACKGROUND: Fulvestrant is a selective oestrogen receptor down-regulator (SERD), which by blocking proliferation of breast cancer cells, is an effective endocrine treatment for women with hormone-sensitive advanced breast cancer. The goal of such systemic therapy in this setting is to reduce symptoms, improve quality of life, and increase survival time. OBJECTIVES: To assess the efficacy and safety of fulvestrant for hormone-sensitive locally advanced or metastatic breast cancer in postmenopausal women, as compared to other standard endocrine agents. SEARCH METHODS: We searched the Cochrane Breast Cancer Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, the World Health Organization International Clinical Trials Registry Platform (WHO ICTRP), and ClinicalTrials.gov on 7 July 2015. We also searched major conference proceedings (American Society of Clinical Oncology (ASCO) and San Antonio Breast Cancer Symposium) and practice guidelines from major oncology groups (ASCO, European Society for Medical Oncology (ESMO), National Comprehensive Cancer Network, and Cancer Care Ontario). We handsearched reference lists from relevant studies. SELECTION CRITERIA: We included for analyses randomised controlled trials that enrolled postmenopausal women with hormone-sensitive advanced breast cancer (TNM classifications: stages IIIA, IIIB, and IIIC) or metastatic breast cancer (TNM classification: stage IV) with an intervention group treated with fulvestrant with or without other standard anticancer therapy. DATA COLLECTION AND ANALYSIS: Two review authors independently extracted data from trials identified in the searches, conducted 'Risk of bias' assessments of the included studies, and assessed the overall quality of the evidence using the GRADE approach. Outcome data extracted from these trials for our analyses and review included progression-free survival (PFS) or time to progression (TTP) or time to treatment failure, overall survival, clinical benefit rate, toxicity, and quality of life. We used the fixed-effect model for meta-analysis where possible. MAIN RESULTS: We included nine studies randomising 4514 women for meta-analysis and review. Overall results for the primary endpoint of PFS indicated that women receiving fulvestrant did at least as well as the control groups (hazard ratio (HR) 0.95, 95% confidence interval (CI) 0.89 to 1.02; P = 0.18, I2= 56%, 4258 women, 9 studies, high-quality evidence). In the one high-quality study that tested fulvestrant at the currently approved and now standard dose of 500 mg against anastrozole, women treated with fulvestrant 500 mg did better than anastrozole, with a HR for TTP of 0.66 (95% CI 0.47 to 0.93; 205 women) and a HR for overall survival of 0.70 (95% CI 0.50 to 0.98; 205 women). There was no difference in PFS whether fulvestrant was used in combination with another endocrine therapy or in the first- or second-line setting, when compared to control treatments: for monotherapy HR 0.97 (95% CI 0.90 to 1.04) versus HR 0.87 (95% CI 0.77 to 0.99) for combination therapy when compared to control, and HR 0.93 (95% CI 0.84 to 1.03) in the first-line setting and HR 0.96 (95% CI 0.88 to 1.04) in the second-line setting.Overall, there was no difference between fulvestrant and control treatments in clinical benefit rate (risk ratio (RR) 1.03, 95% CI 0.97 to 1.10; P = 0.29, I2 = 24%, 4105 women, 9 studies, high-quality evidence) or overall survival (HR 0.97, 95% CI 0.87 to 1.09, P = 0.62, I2 = 66%, 2480 women, 5 studies, high-quality evidence). There was no significant difference in vasomotor toxicity (RR 1.02, 95% CI 0.89 to 1.18, 3544 women, 8 studies, high-quality evidence), arthralgia (RR 0.96, 95% CI 0.86 to 1.09, 3244 women, 7 studies, high-quality evidence), and gynaecological toxicities (RR 1.22, 95% CI 0.94 to 1.57, 2848 women, 6 studies, high-quality evidence). Four studies reported quality of life, none of which reported a difference between the fulvestrant and control arms, though specific data were not presented. AUTHORS' CONCLUSIONS: For postmenopausal women with advanced hormone-sensitive breast cancer, fulvestrant is at least as effective and safe as the comparator endocrine therapies in the included studies. However, fulvestrant may be potentially more effective than current therapies when given at 500 mg, though this higher dosage was used in only one of the nine studies included in the review. We saw no advantage with combination therapy, and fulvestrant was equally as effective as control therapies in both the first- and second-line setting. Our review demonstrates that fulvestrant is a safe and effective systemic therapy and can be considered as a valid option in the sequence of treatments for postmenopausal women with hormone-sensitive advanced breast cancer.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Estradiol/análogos & derivados , Anastrozol , Antineoplásicos Hormonais/efeitos adversos , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Intervalo Livre de Doença , Estradiol/efeitos adversos , Estradiol/uso terapêutico , Feminino , Fulvestranto , Humanos , Neoplasias Hormônio-Dependentes/tratamento farmacológico , Nitrilas/uso terapêutico , Ensaios Clínicos Controlados Aleatórios como Assunto , Triazóis/uso terapêutico
8.
Cancer Prev Res (Phila) ; 10(1): 76-88, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27756753

RESUMO

Over one third of patients who have undergone oral squamous cell carcinoma (OSCC) surgical resections develop life-threatening and often untreatable recurrences. A variety of drugs, intended for management of recurrent or disseminated cancers, were designed to exploit cancer cells' reliance upon overexpressed receptors and gratuitous signaling. Despite their conceptual promise, clinical trials showed these agents lacked efficacy and were often toxic. These findings are consistent with evasion of pathway-targeted treatments via extensive signaling redundancies and compensatory mechanisms common to cancers. Optimal secondary OSCC chemoprevention requires long-term efficacy with multifaceted, nontoxic agents. Accordingly, this study evaluated the abilities of three complementary chemopreventives, that is, the vitamin A derivative fenretinide (4-HPR, induces apoptosis and differentiation, inhibits signaling proteins, and invasion), the estrogen metabolite 2-methoxyestradiol (2-ME, apoptosis-inducing, antiangiogenic), and the humanized mAb to the IL6R receptor tocilizumab (TOC, reduces IL6 signaling) to suppress OSCC gratuitous signaling and tumorigenesis. Modeling studies demonstrated 4-HPR's high-affinity binding at STAT3's dimerization site and c-Abl and c-Src ATP-binding kinase sites. Although individual agents suppressed cancer-promoting pathways including STAT3 phosphorylation, STAT3-DNA binding, and production of the trans-signaling enabling sIL6R, maximal chemopreventive effects were observed with agent combinations. OSCC tumor xenograft studies showed that locally delivered TOC, TOC+4-HPR, and TOC+4-HPR+2-ME treatments all prevented significant tumor growth. Notably, the TOC+4-HPR+2-ME treatment resulted in the smallest overall increase in tumor volume. The selected agents use diverse mechanisms to disrupt tumorigenesis at multiple venues, that is, intracellular, tumor cell-ECM, and tumor microenvironment; beneficial qualities for secondary chemopreventives. Cancer Prev Res; 10(1); 76-88. ©2016 AACR.


Assuntos
Anticarcinógenos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinogênese/efeitos dos fármacos , Carcinoma de Células Escamosas/prevenção & controle , Neoplasias Bucais/prevenção & controle , Recidiva Local de Neoplasia/prevenção & controle , 2-Metoxiestradiol , Animais , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticarcinógenos/administração & dosagem , Anticarcinógenos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/cirurgia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estradiol/análogos & derivados , Estradiol/uso terapêutico , Fenretinida/administração & dosagem , Fenretinida/efeitos adversos , Fenretinida/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Bucais/patologia , Neoplasias Bucais/cirurgia , Invasividade Neoplásica , Fenótipo , Fosforilação , Receptores de Interleucina-6/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Biomed Pharmacother ; 82: 520-7, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27470392

RESUMO

OBJECTIVE: Currently, medicinal plants are found to have biological and pharmacological activities and are used in various domains. This study, carried out on Wistar rats, evaluates the beneficial effects of Artemisia arborscens extract on oestroprogestative-induced damages in kidney. MATERIALS AND METHODS: Thirty-six 3-month-old Wistar rats were divided into 4 batches of nine each: a control group, a group of rats receiving oestroprogestative treatment, a group undergoing oestroprogestative treatment after receiving Artemisia arborescens extract in drinking water, and a group that received only Artemisia arborescens. RESULTS: Artemisia arborescens extract was found to optimize many parameters which were shifted to pathological values as a consequence of oestroprogestative toxicity: plasma creatinine and urea levels were decreased, uric acid and proteins were restored to normal values. The alteration of renal architecture was also suppressed. In addition, superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPX) activities that had been reduced in kidney of the treated group were restored by Aretmisia arborscens-based treatments and, therefore, the lipid peroxidation level was reduced in the renal tissue compared to the control group. CONCLUSION: The obtained results confirmed that the Artemisia-based treatment allowed efficient protection against oestroprogestative-induced nephrotoxicity by restoring the activities of kidney. The protective effect of Artemisia arborescens was mainly attributed to antioxidant properties as well as the presence of phenolic acids and flavonoids detected by means of HPLC.


Assuntos
Antioxidantes/farmacologia , Artemisia/química , Estradiol/efeitos adversos , Rim/patologia , Extratos Vegetais/farmacologia , Progesterona/efeitos adversos , Substâncias Protetoras/farmacologia , Animais , Biomarcadores/sangue , Compostos de Bifenilo/química , Cromatografia Líquida de Alta Pressão , Etanol/química , Sequestradores de Radicais Livres/farmacologia , Rim/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Picratos/química , Ratos Wistar , Água/química
10.
Nutr Neurosci ; 19(10): 467-474, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25087680

RESUMO

OBJECTIVES: Genistein is a plant-derived estrogenic isoflavone commonly found in dietary and therapeutic supplements, due to its potential health benefits. Growth hormone-releasing hormone (GHRH) and somatostatin (SS) are neurosecretory peptides synthesized in neurons of the hypothalamus and regulate the growth hormone secretion. Early reports indicate that estrogens have highly involved in the regulation of GHRH and SS secretions. Since little is known about the potential effects of genistein on GHRH and SS neurons, we exposed rats to genistein. METHODS: Genistein were administered to adult rats in dose of 30 mg/kg, for 3 weeks. The estradiol-dipropionate treatment was used as the adequate controls to genistein. Using applied stereology on histological sections of hypothalamus, we obtained the quantitative information on arcuate (Arc) and periventricular (Pe) nucleus volume and volume density of GHRH neurons and SS neurons. Image analyses were used to obtain GHRH and SS contents in the median eminence (ME). RESULTS: Administration of estradiol-dipropionate caused the increase of Arc and Pe nucleus volume, SS neuron volume density, GHRH and SS staining intensity in the ME, when compared with control. Genistein treatment increased: Arc nucleus volume and the volume density of GHRH neurons (by 26%) and SS neurons (1.5 fold), accompanied by higher GHRH and SS staining intensity in the ME, when compared to the orhidectomized group. DISCUSSION: These results suggest that genistein has a significant effect on hypothalamic region, involved in the regulation of somatotropic system function, and could contribute to the understanding of genistein as substance that alter the hormonal balance.


Assuntos
Genisteína/farmacologia , Hormônio Liberador de Hormônio do Crescimento/agonistas , Hipotálamo/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Fitoestrógenos/farmacologia , Somatostatina/agonistas , Animais , Núcleo Arqueado do Hipotálamo/citologia , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/crescimento & desenvolvimento , Núcleo Arqueado do Hipotálamo/metabolismo , Tamanho Celular/efeitos dos fármacos , Suplementos Nutricionais/efeitos adversos , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estradiol/análogos & derivados , Estradiol/farmacologia , Estrogênios/administração & dosagem , Estrogênios/efeitos adversos , Estrogênios/farmacologia , Genisteína/administração & dosagem , Genisteína/efeitos adversos , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hipotálamo/citologia , Hipotálamo/crescimento & desenvolvimento , Hipotálamo/metabolismo , Injeções Subcutâneas , Masculino , Eminência Mediana/citologia , Eminência Mediana/efeitos dos fármacos , Eminência Mediana/crescimento & desenvolvimento , Eminência Mediana/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Orquiectomia , Tamanho do Órgão/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/crescimento & desenvolvimento , Núcleo Hipotalâmico Paraventricular/metabolismo , Fitoestrógenos/administração & dosagem , Fitoestrógenos/efeitos adversos , Ratos Wistar , Somatostatina/metabolismo , Técnicas Estereotáxicas
11.
Obstet Gynecol ; 126(2): 413-422, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26241433

RESUMO

OBJECTIVE: To describe the effects of six interventions for menopausal vasomotor symptoms relative to control in a pooled analysis, facilitating translation of the results for clinicians and symptomatic women. The Menopause Strategies: Finding Lasting Answers for Symptoms and Health network tested these interventions in three randomized clinical trials. METHODS: An analysis of pooled individual-level data from three randomized clinical trials is presented. Participants were 899 perimenopausal and postmenopausal women with at least 14 bothersome vasomotor symptoms per week. Interventions included 10-20 mg escitalopram per day, nonaerobic yoga, aerobic exercise, 1.8 g per day omega-3 fatty acid supplementation, 0.5 mg low-dose oral 17-beta-estradiol (E2) per day, and 75 mg low-dose venlafaxine XR per day. The main outcome measures were changes from baseline in mean daily vasomotor symptom frequency and bother during 8-12 weeks of treatment. Linear regression models estimated differences in outcomes between each intervention and corresponding control group adjusted for baseline characteristics. Models included trial-specific intercepts, effects of the baseline outcome measure, and time. RESULTS: The 8-week reduction in vasomotor symptom frequency from baseline relative to placebo was similar for escitalopram at -1.4 per day (95% confidence interval [CI] -2.7 to -0.2), low-dose E2 at -2.4 (95% CI -3.4 to -1.3), and venlafaxine at -1.8 (95% CI -2.8 to -0.8); vasomotor symptom bother reduction was minimal and did not vary across these three pharmacologic interventions (mean -0.2 to -0.3 relative to placebo). No effects on vasomotor symptom frequency or bother were seen with aerobic exercise, yoga, or omega-3 supplements. CONCLUSION: These analyses suggest that escitalopram, low-dose E2, and venlafaxine provide comparable, modest reductions in vasomotor symptom frequency and bother among women with moderate hot flushes. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov, www.clinicaltrials.gov, NCT00894543 (MsFLASH 01), NCT01178892 (MsFLASH 02), and NCT01418209 (MsFLASH 03).


Assuntos
Citalopram , Cicloexanóis , Estradiol , Exercício Físico , Ácidos Graxos Ômega-3 , Fogachos , Sistema Vasomotor , Yoga , Citalopram/administração & dosagem , Citalopram/efeitos adversos , Cicloexanóis/administração & dosagem , Cicloexanóis/efeitos adversos , Suplementos Nutricionais , Método Duplo-Cego , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estrogênios/administração & dosagem , Estrogênios/efeitos adversos , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-3/efeitos adversos , Feminino , Fogachos/fisiopatologia , Fogachos/terapia , Humanos , Pessoa de Meia-Idade , Monitorização Fisiológica/métodos , Avaliação de Resultados em Cuidados de Saúde , Perimenopausa/efeitos dos fármacos , Pós-Menopausa/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Inibidores Seletivos de Recaptação de Serotonina/efeitos adversos , Sistema Vasomotor/efeitos dos fármacos , Sistema Vasomotor/fisiopatologia , Cloridrato de Venlafaxina
12.
J Reprod Dev ; 60(4): 274-9, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24769840

RESUMO

Exposure of mice to a high dose of estrogens including diethylstilbestrol (DES) during the neonatal period modifies the developmental plan of the genital tract, which leads to various permanent changes in physiology, morphology and gene expression. These changes include development of an abnormal vaginal epithelium lined with hyperplastic mucinous cells accompanied by Tff1 gene expression in mice. Here, the influence of vitamin D on the direct effect of estrogen on the developing mouse vagina was examined. The mid-vagina of neonatal mice was cultured in a serum-free medium containing estradiol-17ß (E2) and various concentrations of 1,25-dihydroxyvitamin D3 (1,25(OH)2D) ex vivo and then was transplanted under the renal capsule of ovariectomized host mice for 35 days. Exposure to E2 alone caused the vaginal tissue to develop estrogen-independent epithelial hyperplasia and to express TFF1 mRNA, while addition of a low nanomolar amount of 1,25(OH)2D added at the same time as E2 to the culture medium attenuated the effects of estrogen. Expression of vitamin D receptor was also evident in the neonatal mouse vagina. Interestingly, addition of 25-hydroxyvitamin D3, a pro-activated form of vitamin D, at the micromolar level was found to be potent in disrupting the developmental effects of E2, while cholecalciferol was not at least at the dose examined. Correspondingly, expression of Cyp27B1, a kidney-specific 25-hydroxyvitamin D hydroxylase, was evident in the neonatal mouse vagina when examined by RT-PCR. In addition, simultaneous administration of 1,25(OH)2D successfully attenuated DES-induced ovary-independent hyperplasia in the vagina in neonatal mice in vivo. Thus, manipulation of vitamin D influenced the harmful effects of estrogens on mouse vaginal development.


Assuntos
Colecalciferol/farmacologia , Estradiol/efeitos adversos , Estrogênios/efeitos adversos , Vagina/efeitos dos fármacos , Vitaminas/farmacologia , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Animais , Animais Recém-Nascidos , Feminino , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C3H , Peptídeos/metabolismo , Gravidez , Receptores de Calcitriol/metabolismo , Fator Trefoil-1 , Vagina/metabolismo
13.
Clin Drug Investig ; 34(6): 413-20, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24777591

RESUMO

BACKGROUND AND OBJECTIVE: Nomegestrol acetate (NOMAC)/17ß-estradiol (E2) is a monophasic oral contraceptive that contains a progesterone-derived progestogen (NOMAC), and E2, a bio-identical estrogen. The primary objective of this thorough QT/QTc study was to investigate whether once-daily administration of therapeutic (2.5/1.5 mg) and supratherapeutic (12.5/7.5 mg) doses of NOMAC/E2 were associated with prolongation of the mean Fridericia-corrected QT (QTcF) interval in electrocardiograms at steady-state concentrations of NOMAC/E2 versus placebo. The co-primary objective was to establish assay sensitivity after a single dose of moxifloxacin (positive control). METHODS: This was a randomized, double-blind, parallel-group trial comparing 2.5/1.5 mg of NOMAC/E2 (therapeutic dose), 12.5/7.5 mg of NOMAC/E2 (supratherapeutic dose), placebo, and moxifloxacin 400 mg. Double-blind study medication was administered from day -1 to 14. Healthy women aged 18-50 years were randomized. RESULTS: The largest time-matched mean QTcF difference compared with placebo for the therapeutic dose of NOMAC/E2 was 1.6 ms, with an upper limit (UL) of a one-sided 95% confidence interval (CI) of 5.2 ms, and 3.1 ms with an UL 95% CI of 7.0 ms for the supratherapeutic dose. The UL for the time-matched QTcF differences compared with placebo were below the 10 ms threshold defined in the ICH E14 guideline for all time points, both for the therapeutic and the supratherapeutic dose. For moxifloxacin, assay sensitivity was demonstrated. CONCLUSIONS: This thorough QT/QTc study showed that therapeutic and supratherapeutic doses of NOMAC/E2 were not associated with clinically relevant QTc interval prolongation in healthy women after a 2-week period of dosing.


Assuntos
Anticoncepcionais Orais Combinados/efeitos adversos , Estradiol/efeitos adversos , Síndrome do QT Longo/induzido quimicamente , Megestrol/efeitos adversos , Norpregnadienos/efeitos adversos , Administração Oral , Adolescente , Adulto , Anticoncepcionais Orais Combinados/administração & dosagem , Relação Dose-Resposta a Droga , Método Duplo-Cego , Eletrocardiografia , Estradiol/administração & dosagem , Feminino , Fluoroquinolonas/efeitos adversos , Humanos , Megestrol/administração & dosagem , Pessoa de Meia-Idade , Moxifloxacina , Norpregnadienos/administração & dosagem , Fatores de Tempo , Adulto Jovem
14.
Nutr Cancer ; 66(5): 825-34, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24773027

RESUMO

Previous animal studies examining dietary selenium effects on prostatic carcinogenesis did not show preventive benefit, including 1 study in a rat model involving testosterone (T) and estradiol (E2)-induced prostatic oxidative stress. Here, we examined modulation of T + E2-induced prostatic oxidative stress, dysplasia, and inflammation by L-selenomethionine at 1.5 or 3.0 mg selenium/kg in NIH-07 diet in Noble (Nbl)/Crl rats treated with T + E2 for 16 wk. Hormone treatment increased immunohistochemical staining for 8-hydroxydeoxyguanosine (8-OHdG) in the prostatic sites of T + E2-induced preneoplasia (P < 0.05), but selenomethionine did not attenuate 8-OHdG staining and dysplasia in the lateral prostate. Glutathione-peroxidase activity (P < 0.05) and mRNA expression were induced by T + E2 (P < 0.0001) but not changed by selenomethionine. Selenomethionine did not cause significant responses in expression and activity of glutathione-peroxidase and MnSOD, except for a reduction of MnSOD protein expression in the lateral prostate (P < 0.01). The absence of reduction of oxidative stress and dysplasia and the minimal effects on antioxidant enzymes caused by selenomethionine are consistent with the null effects observed in selenium supplementation animal studies and clinical trials. Significant (P < 0.01) opposite apoptosis/cell proliferation balance responses to selenomethionine and to T + E2 occurred in the lateral and dorsal prostate, explaining why T + E2 induces lesions selectively in the lateral lobe of NBL rats.


Assuntos
Estradiol/efeitos adversos , Estresse Oxidativo , Lesões Pré-Cancerosas/prevenção & controle , Próstata/efeitos dos fármacos , Selenometionina/farmacologia , Testosterona/efeitos adversos , 8-Hidroxi-2'-Desoxiguanosina , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Suplementos Nutricionais , Glutationa Peroxidase/metabolismo , Masculino , Lesões Pré-Cancerosas/induzido quimicamente , Próstata/patologia , Substâncias Protetoras/farmacologia , Ratos , Selênio/administração & dosagem , Superóxido Dismutase/metabolismo
15.
Eur J Endocrinol ; 170(1): 121-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24114435

RESUMO

OBJECTIVE: Estrogen stimulates pulsatile secretion of GH, via mechanisms that are largely unknown. An untested hypothesis is that estradiol (E2) drives GH secretion by amplifying interactions among GH-releasing hormone (GHRH), somatostatin (SS), and GH-releasing peptide (GHRP). DESIGN: The design comprised double-blind randomized prospective administration of transdermal E2 vs placebo to healthy postmenopausal women (n=24) followed by pulsatile GHRH or SS infusions for 13 h overnight with or without continuous GHRP2 stimulation. METHODS: End points were mean concentrations, deconvolved secretion, and approximate entropy (ApEn; a regularity measure) of GH. RESULTS: By generalized ANOVA models, it was observed that E2 vs placebo supplementation: i) augmented mean (13-h) GH concentrations (P=0.023), GHRH-induced pulsatile GH secretion over the first 3 h (P=0.0085) and pulsatile GH secretion over the next 10 h (P=0.054); ii) increased GHRP-modulated (P=0.022) and SS-modulated (P<0.001) GH ApEn; and iii) did not amplify GHRH/GHRP synergy during pulsatile GH secretion. By linear regression, E2 concentrations were found to be positively correlated with GH secretion during GHRP2 infusion (P=0.022), whereas BMI was found to be negatively correlated with GH secretion during GHRH (P=0.006) and combined GHRH/GHRP (P=0.015) stimulation. E2 and BMI jointly determined triple (combined l-arginine, GHRH, and GHRP2) stimulation of GH secretion after saline (R²=0.44 and P=0.003) and pulsatile GHRH (R²=0.39 and P=0.013) infusions. CONCLUSION: In summary, in postmenopausal women, E2 supplementation augments the amount (mass) and alters the pattern (regularity) of GH secretion via interactions among GHRH, SS, GHRP, and BMI. These outcomes introduce a more complex model of E2 supplementation in coordinating GH secretion in aging women.


Assuntos
Estradiol/uso terapêutico , Terapia de Reposição de Estrogênios , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Hormônio do Crescimento Humano/metabolismo , Sobrepeso/metabolismo , Adeno-Hipófise/efeitos dos fármacos , Somatostatina/metabolismo , Idoso , Arginina/administração & dosagem , Índice de Massa Corporal , Método Duplo-Cego , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estradiol/farmacocinética , Terapia de Reposição de Estrogênios/efeitos adversos , Retroalimentação Fisiológica/efeitos dos fármacos , Feminino , Hormônio Liberador de Hormônio do Crescimento/administração & dosagem , Hormônio do Crescimento Humano/sangue , Humanos , Infusões Intravenosas , Pessoa de Meia-Idade , Oligopeptídeos/administração & dosagem , Oligopeptídeos/metabolismo , Sobrepeso/sangue , Sobrepeso/fisiopatologia , Adeno-Hipófise/metabolismo , Adeno-Hipófise/fisiopatologia , Pós-Menopausa , Fluxo Pulsátil/efeitos dos fármacos , Somatostatina/administração & dosagem , Adesivo Transdérmico
16.
J Obstet Gynaecol Res ; 39(7): 1259-67, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23803009

RESUMO

AIM: The aim of this study was to investigate the effect of bilateral ovariectomy (OVX), 17-beta estradiol (E2), and progesterone (P4) on the histology and estrogen receptor (ER) expression of the bladder using a female partial bladder outlet obstruction (pBOO) rat model. MATERIAL AND METHODS: A total of 60 female Sprague-Dawley rats were evenly assigned into six groups of 10 each. Group A served as the control. Groups B-F underwent induced pBOO. Groups C-F underwent OVX. Groups D-F were given E2 (0.1 mg/kg/day), Group E was given P4 (1 mg/kg/day), and Group F was given P4 and dehydroepiandrosterone (DHEA) (300 µg/kg/day) by an Alzet pump. Four weeks later, serum E2 and P4 levels were evaluated. Each rat was anesthetized and the urinary bladder was removed for weighing and histological study. RESULTS: Expression of ER-ß was not significantly different between the control group and the other study groups. pBOO was shown to increase both bladder weight and detrusor muscle thickness. OVX had an additive effect to BOO on increased blood vessel density in the bladder. E2 was shown to increase blood vessel density, while P4 supplementation decreased blood vessel density. DHEA did not cause any significant effects on blood vessel density. CONCLUSION: Hormone therapy did not change the expression of ER in bladder outlet obstruction. Estradiol stimulated the increased angiogenesis of the bladder detrusor but P4 decreased the angiogenesis of the bladder detrusor. DHEA had no effect on the bladder detrusor.


Assuntos
Modelos Animais de Doenças , Terapia de Reposição Hormonal , Ovariectomia/efeitos adversos , Receptores de Estrogênio/metabolismo , Obstrução do Colo da Bexiga Urinária/tratamento farmacológico , Bexiga Urinária/efeitos dos fármacos , Indutores da Angiogênese/efeitos adversos , Indutores da Angiogênese/uso terapêutico , Animais , Estradiol/efeitos adversos , Estradiol/uso terapêutico , Feminino , Terapia de Reposição Hormonal/efeitos adversos , Neovascularização Fisiológica/efeitos dos fármacos , Progesterona/efeitos adversos , Progesterona/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Bexiga Urinária/irrigação sanguínea , Bexiga Urinária/metabolismo , Bexiga Urinária/patologia , Obstrução do Colo da Bexiga Urinária/metabolismo , Obstrução do Colo da Bexiga Urinária/patologia
17.
Hum Reprod ; 28(9): 2372-80, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23800622

RESUMO

STUDY QUESTION: What is the effect of estrogen on heparanase procogulant activity? SUMMARY ANSWER: Estrogen increases heparanase procoagulant activity. WHAT IS KNOWN ALREADY: Estrogen therapy increases the risk of thrombosis and was previously found to up-regulate heparanase expression. Heparanase is involved in angiogenesis and metastasis, and has been shown to form a complex with tissue factor (TF) and also shown to enhance the generation of factor Xa. STUDY DESIGN, SIZE, DURATION: A case-control study. Thirty-four healthy women using oral contraceptives (OC) and 41 women not using hormonal therapy and not pregnant per history were enrolled, over a 5-month period, at the Rambam Medical Center, Haifa, Israel. In vitro, estrogen receptor-positive (MCF-7) and -negative (MDA-231) cell lines were incubated with estrogen, tamoxifen and ICI-182.780 a pure estrogen receptor antagonist. The cell medium was evaluated for TF/heparanase complex activity, TF activity and heparanase procoagulant activity by chromogenic substrate. PARTICIPANTS/MATERIALS, SETTING, METHODS: Exclusion criteria included age <18 years, post-menopausal women, concomitant medications other than supplement minerals and vitamins, acute or chronic illness. MAIN RESULTS AND THE ROLE OF CHANCE: The study demonstrates increased risk of high heparanase procoagulant activity in OC users. When a cutoff level of 0.25 (absorbance 405-490 nm) was set, the odds ratio was 131 (P < 0.0001). When all results were studied by quartiles, in quartiles 3 and 4 the results were almost exclusively of the OC users (P < 0.0001). In cell cultures, estrogen and tamoxifen increased heparanase procoagulant activity in the medium of estrogen receptor-positive (MCF-7) cells. LIMITATIONS, REASONS FOR CAUTION: The main limitation of the current study is that the two estrogens given to the women and cell cultures, ethinyl estradiol (EE) and 17-ß-estradiol (E2), respectively, may have different effects on the coagulation system, although an increase in heparanase procoagulant activity was demonstrated in both of them. Although the sample size of the study group was limited, significant differences in the activation of the extrinsic coagulation pathway were demonstrated. WIDER IMPLICATIONS OF THE FINDINGS: The clinical relevance of the heparanase procoagulant activity assay as a screening tool in thrombophilia work-up should further be elucidated.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Anticoncepcionais Orais/efeitos adversos , Glucuronidase/sangue , Trombofilia/induzido quimicamente , Adulto , Estudos de Casos e Controles , Linhagem Celular , Estradiol/efeitos adversos , Estradiol/análogos & derivados , Estradiol/farmacologia , Antagonistas de Estrogênios/farmacologia , Estrogênios/efeitos adversos , Estrogênios/farmacologia , Feminino , Fulvestranto , Glucuronidase/metabolismo , Humanos , Israel/epidemiologia , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/metabolismo , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Risco , Moduladores Seletivos de Receptor Estrogênico/efeitos adversos , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tamoxifeno/efeitos adversos , Tamoxifeno/farmacologia , Trombofilia/sangue , Trombofilia/epidemiologia , Trombofilia/metabolismo , Tromboplastina/metabolismo , Adulto Jovem
18.
Am J Respir Cell Mol Biol ; 49(1): 135-42, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23526212

RESUMO

Lymphangioleiomyomatosis (LAM) is a destructive lung disease primarily affecting women. Genetic studies indicate that LAM cells carry inactivating tuberous sclerosis complex (TSC)-2 mutations, and metastasize to the lung. We previously discovered that estradiol increases the metastasis of TSC2-deficient cells in mice carrying xenograft tumors. Here, we investigate the molecular basis underlying the estradiol-induced lung metastasis of TSC2-deficient cells, and test the efficacy of Faslodex (an estrogen receptor antagonist) in a preclinical model of LAM. We used a xenograft tumor model in which estradiol induces the lung metastasis of TSC2-deficient cells. We analyzed the impact of Faslodex on tumor size, the extracellular matrix organization, the expression of matrix metalloproteinase (MMP)-2, and lung metastasis. We also examined the effects of estradiol and Faslodex on MMP2 expression and activity in tuberin-deficient cells in vitro. Estradiol resulted in a marked reduction of Type IV collagen deposition in xenograft tumors, associated with 2-fold greater MMP2 concentrations compared with placebo-treated mice. Faslodex normalized the Type IV collagen changes in xenograft tumors, enhanced the survival of the mice, and completely blocked lung metastases. In vitro, estradiol enhanced MMP2 transcripts, protein accumulation, and activity. These estradiol-induced changes in MMP2 were blocked by Faslodex. In TSC2-deficient cells, estradiol increased MMP2 concentrations in vitro and in vivo, and induced extracellular matrix remodeling. Faslodex inhibits the estradiol-induced lung metastasis of TSC2-deficient cells. Targeting estrogen receptors with Faslodex may be of efficacy in the treatment of LAM.


Assuntos
Estradiol/análogos & derivados , Estradiol/efeitos adversos , Matriz Extracelular/efeitos dos fármacos , Neoplasias Pulmonares/secundário , Linfangioleiomiomatose/patologia , Remodelação das Vias Aéreas , Animais , Antineoplásicos/farmacologia , Colágeno Tipo IV/metabolismo , Avaliação Pré-Clínica de Medicamentos , Estradiol/farmacologia , Matriz Extracelular/metabolismo , Feminino , Fulvestranto , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Linfangioleiomiomatose/metabolismo , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos SCID , Ratos , Receptores de Estradiol/antagonistas & inibidores , Análise de Sobrevida , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Pediatr Blood Cancer ; 59(1): 27-33, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22162424

RESUMO

BACKGROUND: A new generation of anticancer drugs has reached clinical care in common diseases, but their use in rare diseases such as pediatric brain tumors lags behind since conventional clinical trial design requires larger patient numbers. PROCEDURE: We designed individualized treatment protocols for pediatric patients with relapsed brain tumors, based upon the patient's treatment history. In addition, each tumor was analyzed with morphoproteomics using a panel of markers to show treatment targets, resulting in a list of potential novel drugs to be added to chemotherapy. Here, we present the concept and report the experiences of the first patients enrolled in the program. RESULTS: Eleven treatment protocols were designed using morphoproteomic information and given to eight patients. The histological diagnoses included: medulloblastoma (n = 3), glioblastoma multiforme (n = 2), atypical teratoid rhabdoid tumor (n = 1), choroid plexus carcinoma (n = 1), and primitive neuroectodermal tumors (n = 1). Tumor markers included p-ERK, Topoisomerase IIa, Bcl-2, VEGF-A, p-STAT3, ER-beta, p-mTOR, and p-NF-kappaBp65. The novel agents included sorafenib, bevacizumab, fulvestrant, rapamycin, bortezomib, and curcumin. The response to the first protocol was complete response: 1, partial response: 1, stable disease: 0, progressive disease: 4, and continuous complete remission: 2. The median Event-Free Survival was 0.32 year ± 0.4. For the comparison with the institutional control group, the individual response probability was calculated. The observed response was superior to the historical controls (P = 0.006 Whitman U-test). CONCLUSION: This approach warrants further, systematic evaluation as proof of concept and then expansion to drug-specific hypotheses.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas , Adolescente , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Benzenossulfonatos/administração & dosagem , Benzenossulfonatos/efeitos adversos , Bevacizumab , Ácidos Borônicos/administração & dosagem , Ácidos Borônicos/efeitos adversos , Bortezomib , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Criança , Pré-Escolar , Curcumina/administração & dosagem , Curcumina/efeitos adversos , Intervalo Livre de Doença , Estradiol/administração & dosagem , Estradiol/efeitos adversos , Estradiol/análogos & derivados , Feminino , Seguimentos , Fulvestranto , Humanos , Lactente , Masculino , Niacinamida/análogos & derivados , Compostos de Fenilureia , Pirazinas/administração & dosagem , Pirazinas/efeitos adversos , Piridinas/administração & dosagem , Piridinas/efeitos adversos , Sirolimo/administração & dosagem , Sirolimo/efeitos adversos , Sorafenibe , Taxa de Sobrevida
20.
Physiol Behav ; 105(2): 460-9, 2012 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-21925526

RESUMO

Since estradiol exercises inhibitory effect on food intake, we wanted to find out if this influence of estradiol is mediated by cocaine- and amphetamine-regulated transcript peptide (CART), a well established anorectic agent in the brain. Ovariectomized (OVX) rats, replaced with estradiol to produce estrous-phase like conditions, showed a significant decrease in food intake as compared with that in OVX controls. Intracerebroventricular (icv) administration of CART (0.5-1 µg/rat) to OVX rats, resulted in a dose-dependent reduction in the food intake. The lower dose (0.25 µg) had no effect, and was considered subeffective. In estradiol replaced OVX rats, CART at subeffective dose, further reduced food intake. However, CART failed to reduce food intake in estradiol replaced OVX rats pretreated with anti-estrogenic agent tamoxifen (3 mg/kg, subcutaneous). Administration of CART antibody (1:500 dilution/rat, i.c.v.) significantly attenuated estradiol-induced anorexia in the OVX rats. While estradiol replacement significantly increased CART-immunoreactivity in the cells/fibers of paraventricular nucleus (PVN) of OVX rats, fibers in the anteroventral periventricular nucleus (AVPV), and cells/fibers in the arcuate nucleus (ARC) showed considerable reduction. These changes were attenuated following concurrent injection of tamoxifen to the estradiol replaced OVX rats. However, CART-immunoreactive cells/fibers in the periventricular area did not respond to any of the treatments. We suggest that estradiol treatment might influence the hypothalamic CART system in a site specific manner. While increased CART activity in the PVN might produce anorexia, reduction of CART in ARC and AVPV might represent a compensatory homeostatic response.


Assuntos
Anorexia/induzido quimicamente , Estradiol/efeitos adversos , Estrogênios/efeitos adversos , Proteínas do Tecido Nervoso/metabolismo , Análise de Variância , Animais , Anorexia/tratamento farmacológico , Anorexia/patologia , Anticorpos/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Vias de Administração de Medicamentos , Interações Medicamentosas , Ingestão de Alimentos/efeitos dos fármacos , Antagonistas de Estrogênios/administração & dosagem , Feminino , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/administração & dosagem , Proteínas do Tecido Nervoso/efeitos adversos , Proteínas do Tecido Nervoso/imunologia , Ovariectomia , Ratos , Ratos Sprague-Dawley , Tamoxifeno/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA