Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Transl Med ; 21(1): 232, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-37004031

RESUMO

BACKGROUND: The incidence of obesity, a known risk factor for several metabolic and chronic diseases, including numerous malignancies, has risen sharply in the world. Various clinical studies demonstrate that excessive Body Mass Index (BMI) may worsen the incidence, prognosis, and mortality rates of breast cancer. Thus, understanding the link tying up obesity and breast cancer onset and progression is critically important, as it can impact patients' survival and quality of life. Recently, circulating extracellular vesicle (EV) derived miRNAs have attracted much attention for their diagnostic, prognostic and therapeutic potential in oncology research. Although the potential role of EV-derived miRNAs in the early detection of breast cancer has been repeatedly mentioned, screening of miRNAs packaged within serum EVs has not yet been reported in patients with obesity. METHODS: Circulating EVs were isolated from normal weight (NW), and overweight/obese (OW/Ob) breast cancer patients and characterized by Transmission Electron Microscopy (TEM), Nanoparticle Tracking Analysis (NTA), and protein marker expression. Evaluation of EV-associated miRNAs was conducted in a screening (RNA-seq) and a validation (qRT-PCR) cohort. Bioinformatic analysis was performed to uncover significantly enriched biological processes, molecular functions and pathways. ROC and Kaplain-Meier survival analyses were used for clinical significance. RESULTS: Comparison of serum EV-derived miRNAs from NW and OW/Ob patients detected seven differentially expressed miRNAs (let-7a-5p, miR-122-5p, miR-30d-5p, miR-126-3p, miR-27b-3p, miR-4772-3p, and miR-10a-5p) in the screening cohort. GO analysis revealed the enrichment of protein phosphorylation, intracellular signal transduction, signal transduction, and vesicle-mediated transport among the top biological processes. In addition, the target genes were significantly enriched in pathways related to PI3K/Akt, growth hormones, and insulin signalings, which are all involved in obesity-related diseases and/or breast cancer progression. In the validation cohort, qRT-PCR confirmed a significant down-regulation of EV-derived let-7a in the serum of OW/Ob breast cancer patients compared to NW patients. Let-7a levels also exhibited a negative correlation with BMI values. Importantly, decreased let-7a miRNA expression was associated with higher tumor grade and poor survival in patients with breast cancer. CONCLUSION: These results suggest that serum-EV derived miRNAs may reflect a differential profile in relation to a patient's BMI, which, once validated in larger cohorts of patients, could provide insights into novel specific biomarkers and innovative targets to prevent the progression of obesity-mediated breast cancer.


Assuntos
Neoplasias da Mama , MicroRNA Circulante , Vesículas Extracelulares , MicroRNAs , Humanos , Feminino , MicroRNA Circulante/metabolismo , Neoplasias da Mama/complicações , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Qualidade de Vida , MicroRNAs/metabolismo , Vesículas Extracelulares/metabolismo , Obesidade/complicações , Obesidade/genética , Obesidade/metabolismo
2.
Int J Mol Sci ; 24(17)2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37686282

RESUMO

Androgen receptor (AR) expression in estrogen receptor-positive (ER+) breast cancer (BC) correlates with lower tumor grade and a better clinical outcome. Additionally, in normal mammary epithelium or ER+ BC preclinical models, androgens counteract basal/ER-dependent proliferation. Here, we report an additional mechanism, underlining the protective role exerted by AR. Specifically, the activation of intracellular AR upregulates the Bcl-2-family protein BAD, and TCGA database analyses show that in ER+ BC, BAD expression is associated with better disease-free survival. Ligand-activated AR influences its own and BAD cellular compartmentalization by enhancing levels in the nucleus, as well as in mitochondrial fractions. In both compartments, BAD exerts unconventional functions. In the nucleus, BAD and AR physically interact and, upon androgen stimulation, are recruited at the AP-1 and ARE sites within the cyclin D1 promoter region, contributing to explaining the anti-proliferative effect of androgens in BC cells. Androgens cause an enrichment in BAD and AR content in the mitochondria, correlated with a decrease in mitochondrial function. Thus, we have defined a novel mechanism by which androgens modulate BAD expression, its mitochondria localization, and nuclear content to force its ability to act as a cell cycle inhibitor, strengthening the protective role of androgen signaling in estrogen-responsive BCs.


Assuntos
Androgênios , Neoplasias , Androgênios/farmacologia , Morte Celular , Núcleo Celular , Estrogênios , Ciclo Celular
3.
Int J Mol Sci ; 24(21)2023 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-37958677

RESUMO

Circulating extracellular vesicle (EV)-derived microRNAs (miRNAs) are now considered the next generation of cancer "theranostic" tools, with strong clinical relevance. Although their potential in breast cancer diagnosis has been widely reported, further studies are still required to address this challenging issue. The present study examined the expression profiles of EV-packaged miRNAs to identify novel miRNA signatures in breast cancer and verified their diagnostic accuracy. Circulating EVs were isolated from healthy controls and breast cancer patients and characterized following the MISEV 2018 guidelines. RNA-sequencing and real-time PCR showed that miRNA-27a and miRNA-128 were significantly down-regulated in patient-derived EVs compared to controls in screening and validation cohorts. Bioinformatics analyses of miRNA-target genes indicated several enriched biological processes/pathways related to breast cancer. Receiver operating characteristic (ROC) curves highlighted the ability of these EV-miRNAs to distinguish breast cancer patients from non-cancer controls. According to other reports, the levels of EV-miRNA-27a and EV-miRNA-128 are not associated with their circulating ones. Finally, evidence from the studies included in our systematic review underscores how the expression of these miRNAs in biofluids is still underinvestigated. Our findings unraveled the role of serum EV-derived miRNA-27a and miRNA-128 in breast cancer, encouraging further investigation of these two miRNAs within EVs towards improved breast cancer detection.


Assuntos
Neoplasias da Mama , Vesículas Extracelulares , MicroRNAs , Humanos , Feminino , MicroRNAs/metabolismo , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo
4.
J Transl Med ; 20(1): 107, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35241092

RESUMO

BACKGROUND: Plasma lipid profile and anthropometric variables are known to be under strong genetic control and the identification of genetic variants associated with bioclinical parameters is of considerable public health importance. In this study, a young cohort of healthy individuals was genotyped for genes related to health and pathological conditions, to analyze the association of single nucleotide polymorphisms (SNPs) with different bioclinical parameters, adherence to the Mediterranean Diet (MD) and physical activity, studying the role of lifestyle and body composition parameters on biochemical metabolic profile. METHODS: Association analysis of single variants in the genes of lipoprotein lipase (LPL), fibronectin type III domain containing protein 5 (FNDC5), and peroxisome proliferator-activated receptor-gamma (PPARγ) and haplotype analyses were performed. RESULTS: Multiple (n = 14) common variants in the three genes demonstrated a significant effect on plasma lipoprotein-lipid levels and/or on biochemical parameters in our sample. Specifically, SNPs were related to lipid metabolism (rs3866471, rs4922115, rs11570892, rs248, rs316, rs1059507, rs1801282) or glycemic profile (rs3208305) or anthropometric parameters (rs3480, rs726344, rs1570569) for a total of 26 significant associations (P < 0.01 and/or P < 0.05) and two haplotypes, for the first time, were strongly associated with lipid and body composition parameters. Interestingly, we identified twenty-four new variants not previously described in the literature and a novel significant association between rs80143795 and body composition. CONCLUSIONS: In this study we confirm the association between these SNPs on lipid metabolism and body parameters also in a young cohort, indicating the important role of these genetic factors as determinants of health.


Assuntos
Lipase Lipoproteica , PPAR gama , Adolescente , Composição Corporal/genética , Fibronectinas/genética , Humanos , Lipídeos , Lipase Lipoproteica/genética , Metaboloma , PPAR gama/genética , Polimorfismo de Nucleotídeo Único/genética
5.
Int J Mol Sci ; 23(21)2022 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-36361728

RESUMO

Tumor extracellular vesicles (EVs), as endocytic vesicles able to transport nucleic acids, proteins, and metabolites in recipient cells, have been recognized fundamental mediators of cell-to-cell communication in breast cancer. The biogenesis and release of EVs are highly regulated processes and both the quantity of EVs and their molecular cargo might reflect the metabolic state of the producing cells. We recently demonstrated that the adipokine leptin, whose circulating levels correlate with adipose tissue expansion, is an inducer of EV release from breast cancer cells. Here, we show a specific proteomic signature of EVs released by MCF-7 breast cancer cells grown in the presence of leptin (Lep-EVs), in attempt to find additional molecular effectors linking obesity to breast cancer biology. An analysis of the proteomic profile of Lep-EVs by LC-MS/MS revealed a significant enrichment in biological processes, molecular functions, and cellular components mainly related to mitochondrial machineries and activity, compared to protein content of EVs from untreated breast cancer cells. Metabolic investigations, carried out to assess the autocrine effects of these vesicles on breast cancer cells, revealed that Lep-EVs were able to increase ATP levels in breast cancer cells. This result is associated with increased mitochondrial respiration evaluated by Seahorse analyzer, supporting the concept that Lep-EVs can modulate MCF-7 breast cancer cell oxidative metabolism. Moreover, taking into account the relevance of tumor immune cell crosstalk in the tumor microenvironment (TME), we analyzed the impact of these vesicles on macrophage polarization, the most abundant immune component in the breast TME. We found that tumor-derived Lep-EVs sustain the polarization of M0 macrophages, derived from the human THP-1 monocytic cells, into M2-like tumor-associated macrophages, in terms of metabolic features, phagocytic activity, and increased expression of CD206-positive population. Overall, our results indicate that leptin by inducing the release of EV-enriched in mitochondrial proteins may control the metabolism of MCF-7 breast cancer cells as well as that of macrophages. Characterization of tumor-derived EV protein cargo in an obesity-associated milieu, such as in the presence of elevated leptin levels, might allow identifying unique features and specific metabolic mechanisms useful to develop novel therapeutic approaches for treatment of breast cancer, especially in obese patients.


Assuntos
Neoplasias da Mama , Vesículas Extracelulares , Humanos , Feminino , Proteômica , Neoplasias da Mama/metabolismo , Leptina/metabolismo , Cromatografia Líquida , Espectrometria de Massas em Tandem , Vesículas Extracelulares/metabolismo , Obesidade/metabolismo , Microambiente Tumoral
6.
Semin Cancer Biol ; 60: 274-284, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31491560

RESUMO

The escalating epidemic of overweight and obesity is currently recognized as one of the most significant health and economic concern worldwide. At the present time, over 1.9 billion adults and more than 600 million people can be, respectively, classified as overweight or obese, and numbers will continue to increase in the coming decades. This alarming scenario implies important clinical implications since excessive adiposity can progressively cause and/or exacerbate a wide spectrum of co-morbidities, including type 2 diabetes mellitus, hypertension, cardiovascular disease, and even certain types of cancer, including breast cancer. Indeed, pathological remodelling of white adipose tissue and increased levels of fat-specific cytokines (mainly leptin), as a consequence of the obesity condition, have been associated with several hallmarks of breast cancer, such as sustained proliferative signaling, cellular energetics, inflammation, angiogenesis, activating invasion and metastasis. Different preclinical and clinical data have provided evidence indicating that obesity may worsen the incidence, the severity, and the mortality of breast cancer. In the present review, we will discuss the epidemiological connection between obesity and breast cancer progression and metastasis and we will highlight the candidate players involved in this dangerous relationship. Since the major cause of death from cancer is due to widespread metastases, understanding these complex mechanisms will provide insights for establishing new therapeutic interventions to prevent/blunt the effects of obesity and thwart breast tumor progression and metastatic growth.


Assuntos
Neoplasias da Mama/complicações , Neoplasias da Mama/patologia , Obesidade/complicações , Biomarcadores , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/metabolismo , Progressão da Doença , Transição Epitelial-Mesenquimal , Feminino , Humanos , Leptina/metabolismo , Metástase Neoplásica , Estadiamento de Neoplasias , Neovascularização Patológica
7.
Am J Pathol ; 189(3): 687-698, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30610844

RESUMO

Although in past decades the adipokine leptin and its own receptor have been considered as significant cancer biomarkers, their potential involvement in human testicular seminoma growth and progression remains unexplored. Here, we showed that the expression of leptin and its receptor was significantly higher in human testicular seminoma compared with normal adult testis. Human seminoma cell line TCam-2 also expressed leptin along with the long and short isoforms of leptin receptor, and in response to leptin treatment showed enhanced activation of its downstream effectors. In line with these results, leptin stimulation significantly increased the proliferation and migration of TCam-2 cells. Treatment of TCam-2 cells with the peptide Leu-Asp-Phe-Ile (LDFI), a full leptin-receptor antagonist, completely reversed the leptin-mediated effects on cell growth and motility as well as reduced the expression of several leptin-induced target genes. More importantly, the in vivo xenograft experiments showed that LDFI treatment markedly decreased seminoma tumor growth. Interestingly, LDFI-treated tumors showed reduced levels of the proliferation marker Ki-67 as well as decreased expression of leptin-regulated genes. Taken together, these data identify, for the first time, leptin as a key factor able to affect testicular seminoma behavior, highlighting leptin receptor as a potential target for novel potential treatments in this type of cancer.


Assuntos
Leptina/farmacocinética , Proteínas de Neoplasias/agonistas , Peptídeos/farmacologia , Receptores para Leptina/agonistas , Seminoma/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico , Adulto , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Leptina/química , Masculino , Camundongos , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Peptídeos/química , Receptores para Leptina/metabolismo , Seminoma/metabolismo , Seminoma/patologia , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Int J Food Sci Nutr ; 71(5): 581-592, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31690142

RESUMO

In this cross-sectional web-survey, carried out in 340 employees (24-67 years) among university staff of Southern Italy, we assessed the adherence to the Mediterranean Diet (MD). Using an online questionnaire based on validated 14-point MD Adherence Screener (MEDAS), the mean of the score was 7.34 (±1.9) for total population independently of sex. In population divided by the cut-off age of 45 years, MD adherence resulted significantly lower in younger respect to older group (p = .003). In multiple regression analyses we observed the direct association between MEDAS score and older age group also after adjustments. Importantly, in all sample the percentage of adherers to recommendations for fruits, nuts and fish resulted outside dietary guidelines. The present findings underscore the need to develop healthy education programmes aimed to improve the consumption of several components of the MD, particularly among young adults, in order to prevent the early onset of chronic non-transmittable diseases.


Assuntos
Dieta Mediterrânea , Comportamento Alimentar , Comportamentos Relacionados com a Saúde , Universidades , Adulto , Fatores Etários , Idoso , Estudos Transversais , Inquéritos sobre Dietas , Feminino , Humanos , Internet , Itália/epidemiologia , Masculino , Pessoa de Meia-Idade , Política Nutricional , Adulto Jovem
9.
Int J Mol Sci ; 21(7)2020 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-32224850

RESUMO

Over the last decades a renewed interest in n-3 very long polyunsaturated fatty acids (PUFAs), derived mainly from fish oils in the human diet, has been observed because of their potential effects against cancer diseases, including breast carcinoma. These n-3 PUFAs mainly consist of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) that, alone or in combination with anticancer agents, induce cell cycle arrest, autophagy, apoptosis, and tumor growth inhibition. A large number of molecular targets of n-3 PUFAs have been identified and multiple mechanisms appear to underlie their antineoplastic activities. Evidence exists that EPA and DHA also elicit anticancer effects by the conversion to their corresponding ethanolamide derivatives in cancer cells, by binding and activation of different receptors and distinct signaling pathways. Other conjugates with serotonin or dopamine have been found to exert anti-inflammatory activities in breast tumor microenvironment, indicating the importance of these compounds as modulators of tumor epithelial/stroma interplay. The objective of this review is to provide a general overview and an update of the current n-3 PUFA derivative research and to highlight intriguing aspects of the potential therapeutic benefits of these low-toxicity compounds in breast cancer treatment and care.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/prevenção & controle , Ácidos Docosa-Hexaenoicos/farmacologia , Ácido Eicosapentaenoico/farmacologia , Animais , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/uso terapêutico , Ácido Eicosapentaenoico/uso terapêutico , Feminino , Humanos , Microambiente Tumoral/efeitos dos fármacos
10.
Int J Mol Sci ; 21(24)2020 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-33352766

RESUMO

Multiple lines of evidence indicate that activation of the peroxisome proliferator-activated receptor γ (PPARγ) by natural or synthetic ligands exerts tumor suppressive effects in different types of cancer, including breast carcinoma. Over the past decades a new picture of breast cancer as a complex disease consisting of neoplastic epithelial cells and surrounding stroma named the tumor microenvironment (TME) has emerged. Indeed, TME is now recognized as a pivotal element for breast cancer development and progression. Novel strategies targeting both epithelial and stromal components are under development or undergoing clinical trials. In this context, the aim of the present review is to summarize PPARγ activity in breast TME focusing on the role of this receptor on both epithelial/stromal cells and extracellular matrix components of the breast cancer microenvironment. The information provided from the in vitro and in vivo research indicates PPARγ ligands as potential agents with regards to the battle against breast cancer.


Assuntos
Neoplasias da Mama/patologia , PPAR gama/metabolismo , Microambiente Tumoral/imunologia , Animais , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Feminino , Humanos , Ligantes , Transdução de Sinais
11.
Int J Mol Sci ; 21(16)2020 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-32823947

RESUMO

Aromatase inhibitors (AIs) represent the standard anti-hormonal therapy for post-menopausal estrogen receptor-positive breast cancer, but their efficacy is limited by the emergence of AI resistance (AIR). Exosomes act as vehicles to engender cancer progression and drug resistance. The goal of this work was to study exosome contribution in AIR mechanisms, using estrogen-dependent MCF-7 breast cancer cells as models and MCF-7 LTED (Long-Term Estrogen Deprived) subline, modeling AIR. We found that exosome secretion was significantly increased in MCF-7 LTED cells compared to MCF-7 cells. MCF-7 LTED cells also exhibited a higher amount of exosomal RNA and proteins than MCF-7 cells. Proteomic analysis revealed significant alterations in the cellular proteome. Indeed, we showed an enrichment of proteins frequently identified in exosomes in MCF-7 LTED cells. The most up-regulated proteins in MCF-7 LTED cells were represented by Rab GTPases, important vesicle transport-regulators in cancer, that are significantly mapped in "small GTPase-mediated signal transduction", "protein transport" and "vesicle-mediated transport" Gene Ontology categories. Expression of selected Rab GTPases was validated by immunoblotting. Collectively, we evidence, for the first time, that AIR breast cancer cells display an increased capability to release exosomes, which may be associated with an enhanced Rab GTPase expression. These data provide the rationale for further studies directed at clarifying exosome's role on endocrine therapy, with the aim to offer relevant markers and druggable therapeutic targets for the management of hormone-resistant breast cancers.


Assuntos
Inibidores da Aromatase/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Exossomos/metabolismo , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Análise por Conglomerados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Estrogênios/deficiência , Exossomos/ultraestrutura , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Proteômica , Regulação para Cima/efeitos dos fármacos , Proteínas rab de Ligação ao GTP/metabolismo
12.
Int J Mol Sci ; 20(9)2019 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-31052147

RESUMO

Obesity, characterized by excess body weight, is now accepted as a hazardous health condition and an oncogenic factor. In different epidemiological studies obesity has been described as a risk factor in several malignancies. Some biological mechanisms that orchestrate obesity-cancer interaction have been discovered, although others are still not completely understood. The unbalanced secretion of biomolecules, called "adipokines", released by adipocytes strongly influences obesity-related cancer development. Among these adipokines, adiponectin exerts a critical role. Physiologically adiponectin governs glucose levels and lipid metabolism and is fundamental in the reproductive system. Low adiponectin circulating levels have been found in obese patients, in which its protective effects were lost. In this review, we summarize the epidemiological, in vivo and in vitro data in order to highlight how adiponectin may affect obesity-associated female cancers.


Assuntos
Adiponectina/metabolismo , Neoplasias da Mama/metabolismo , Obesidade/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias da Mama/epidemiologia , Feminino , Humanos , Obesidade/epidemiologia , Neoplasias Ovarianas/epidemiologia , Transdução de Sinais
13.
Am J Pathol ; 186(5): 1328-39, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26968343

RESUMO

Leydig cell tumors are the most frequent interstitial neoplasms of the testis with increased incidence in recent years. They are hormonally active and are considered one of the steroid-secreting tumors. Although usually benign, the malignant phenotype responds poorly to conventional chemotherapy or radiation, highlighting the need to identify new therapeutic targets for treatment. Here, we identified a novel glucocorticoid-mediated mechanism that controls cell growth in Leydig cell tumors. We found that a synthetic glucocorticoid receptor agonist, dexamethasone, reduces cell proliferation in rat Leydig tumor cells by decreasing the expression and the enzymatic activity of the estrogen-producing enzyme aromatase. This inhibitory effect relies on the ability of activated glucocorticoid receptor to regulate the aromatase gene transcriptional activity through the recruitment of nuclear receptor corepressor protein and silencing mediator of retinoid and thyroid hormone receptors to a newly identified putative glucocorticoid responsive element within the aromatase promoter II. Our in vivo studies reveal a reduction of tumor growth, after dexamethasone treatment, in animal xenografts. Tumors from dexamethasone-treated mice exhibit a decrease in the expression of the proliferation marker Ki-67 and the aromatase enzyme. Our data demonstrate that activated glucocorticoid receptor, decreasing aromatase expression, induces Leydig tumor regression both in vitro and in vivo, suggesting that glucocorticoid receptor might be a potential target for the therapy of Leydig cell tumors.


Assuntos
Inibidores da Aromatase/farmacologia , Aromatase/metabolismo , Dexametasona/farmacologia , Tumor de Células de Leydig/patologia , Receptores de Glucocorticoides/antagonistas & inibidores , Neoplasias Testiculares/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Xenoenxertos , Tumor de Células de Leydig/tratamento farmacológico , Masculino , Camundongos Nus , Transplante de Neoplasias , Neoplasias Testiculares/tratamento farmacológico
14.
Br J Nutr ; 117(1): 170-175, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28098046

RESUMO

I prophylaxis is the most effective strategy to eradicate I deficiency disorders, but it has been shown to affect the thyroid disease pattern. In this study, we assessed the frequency of thyroid disorders in an adult population living in two areas of southern Italy after implementing I prophylaxis. To this aim, a cross-sectional, population-based study including 489 subjects from an I-deficient rural and an I-sufficient urban area of southern Italy was conducted. Thyroid ultrasound was performed on all participants, and urine and blood samples were collected from each subject. The levels of thyroid-stimulating hormone (TSH), thyroglobulin (TgAb) and thyroperoxidase antibodies (TPOAb), urinary I excretion (UIE), and thyroid volume and echogenicity were evaluated. We found that the median UIE was higher in the urban than in the rural area (P=0·004), whereas the prevalence of subjects affected by goitre was higher in the rural compared with the urban area (P=0·003). Positive TgAb rather than TPOAb were more frequent in subjects from the urban area compared with the rural area (P=0·009). The hypoechoic pattern at thyroid ultrasound (HT-US) was similar between the two areas, but TgAb were significantly higher (P=0·01) in HT-US subjects from the urban area. The frequency of elevated TSH did not differ between the two screened populations, and no changes were found for TgAb positivity in subjects with high TSH in the urban compared with the rural area. Our findings support that the small risks of I supplementation are far outweighed by the substantial benefits of correcting I deficiency, although continued monitoring of populations is necessary.


Assuntos
Iodo/administração & dosagem , Iodo/deficiência , Doenças da Glândula Tireoide/epidemiologia , Doenças da Glândula Tireoide/prevenção & controle , Adulto , Estudos Transversais , Feminino , Humanos , Itália , Masculino , Pessoa de Meia-Idade
15.
Biochim Biophys Acta ; 1850(11): 2185-95, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26272430

RESUMO

BACKGROUND: The omega-3 docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) may form conjugates with amines that have potential health benefits against common diseases including cancers. Here we synthesized DHA-dopamine (DHADA) and EPA-dopamine (EPADA) conjugates and studied their biological effects on different breast cancer cell lines. METHODS AND RESULTS: MTT assays indicated that increasing concentrations of DHADA and EPADA significantly affected viability in MCF-7, SKBR3 and MDA-MB-231 breast cancer cells, whereas no effect was observed in MCF-10A non-tumorigenic epithelial breast cells. DHADA and EPADA enhanced Beclin-1 expression, as evidenced by immunoblotting, real-time-PCR and functional analyses. Chromatin Immunoprecipitation (ChIP) and Re-ChIP assays revealed that both compounds induced recruitment of Peroxisome-Proliferator-Activated-Receptor gamma (PPARγ) and RNA Polymerase-II at the Retinoic-X-Receptor binding region on Beclin-1 promoter. Moreover, both compounds enhanced autophagosome formation, evaluated by LC-3 and monodansylcadaverine labeling, that was prevented by the PPARγ antagonist GW9662, addressing the direct involvement of PPARγ. Noteworthy, long-term treatment with DHADA and EPADA caused the blockade of autophagic flux followed by apoptotic cell death as evidenced by PARP cleavage and DNA fragmentation in all breast cancer cells. CONCLUSIONS: We have provided new insights into the molecular mechanism through which PPARγ, as a central molecule in the cross talk between autophagy and apoptosis, mediates DHADA- and EPADA-induced cell death in breast cancer cells. GENERAL SIGNIFICANCE: Our findings suggest that omega-3 DHADA- and EPADA activation of PPARγ may assume biological relevance in setting novel adjuvant therapeutic interventions in breast carcinoma.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/farmacologia , Dopamina/farmacologia , Ácido Eicosapentaenoico/farmacologia , PPAR gama/fisiologia , Proteínas Reguladoras de Apoptose/genética , Proteína Beclina-1 , Neoplasias da Mama/patologia , Feminino , Humanos , Células MCF-7 , Proteínas de Membrana/genética , Regiões Promotoras Genéticas
16.
Chemistry ; 22(9): 3053-64, 2016 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-26821986

RESUMO

The reactivity of 2-(2-alkynylphenoxy)anilines under PdI2 /KI-catalyzed oxidative carbonylation conditions has been studied. Although a different reaction pathway could have been operating, N-palladation followed by CO insertion was the favored pathway with all substrates tested, including those containing an internal or terminal triple bond. This led to the formation of a carbamoylpalladium species, the fate of which, as predicted by theoretical calculations, strongly depended on the nature of the substituent on the triple bond. In particular, 8-endo-dig cyclization preferentially occurred when the triple bond was terminal, leading to the formation of carbonylated ζ-lactam derivatives, the structures of which have been confirmed by XRD analysis. These novel medium-sized heterocyclic compounds showed antitumor activity against both estrogen receptor-positive (MCF-7) and triple negative (MDA-MB-231) breast cancer cell lines. In particular, ζ-lactam 3 j' may represent a novel and promising antitumor agent because biological tests clearly demonstrate that this compound significantly reduces cell viability and motility in both MCF-7 and MDA-MB-231 breast cancer cell lines, without affecting normal breast epithelial cell viability.


Assuntos
Compostos de Anilina/síntese química , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Paládio/química , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Catálise , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclização , Humanos , Lactamas , Oxirredução
17.
FASEB J ; 29(5): 2150-60, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25657113

RESUMO

Obesity is a risk factor for breast cancer, largely due to altered expression of various adipocytokines. As it concerns adiponectin, there are not univocal results regarding its role in breast cancer occurrence and progression. Here, we demonstrate that in animals injected with human estrogen receptor (ER)-α-negative MDA-MB-231 cells pretreated with adiponectin (1 and 5 µg/ml), a significant reduction (60 and 40%, respectively) in tumor volume is observed, whereas an increased tumor growth (54 and 109%, respectively) is evidenced in the animals receiving human ER-α-positive MCF-7 cells. Moreover, cyclin D1 (CD1) mRNA and protein levels are decreased in MDA-MB-231 cells, whereas they are up-regulated in ER-α-positive cells by adiponectin. These findings fit with the opposite effects of adiponectin on CD1 promoter: 0.44- and 0.34-fold decrease in MDA-MB-231 cells and 0.63- and 0.95-fold increase in MCF-7 cells, treated with 1 and 5 µg/ml, respectively. Functional studies indicate that these effects are mediated by the specific protein 1 motif located in the CD1 promoter. In the absence of ER-α, the adiponectin-mediated down-regulation of CD1 involves the recruitment of corepressors. In the presence of ER-α, the adiponectin-induced expression of CD1 requires the involvement of an activator complex. In conclusion, we propose that a possible mechanism through which adiponectin differently affects breast cancer growth is the opposite modulation of CD1 levels accordingly to ER-α expression.


Assuntos
Adiponectina/farmacologia , Neoplasias da Mama/metabolismo , Ciclina D1/metabolismo , Receptor alfa de Estrogênio/metabolismo , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proliferação de Células , Imunoprecipitação da Cromatina , Ciclina D1/genética , Ensaio de Desvio de Mobilidade Eletroforética , Receptor alfa de Estrogênio/genética , Feminino , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Mutagênese Sítio-Dirigida , Mutação/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Células Tumorais Cultivadas
18.
J Cell Mol Med ; 19(5): 1122-32, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25721149

RESUMO

The role of the obesity cytokine leptin in breast cancer progression has raised interest in interfering with leptin's actions as a valuable therapeutic strategy. Leptin interacts with its receptor through three different binding sites: I-III. Site I is crucial for the formation of an active leptin-leptin receptor complex and in its subsequent activation. Amino acids 39-42 (Leu-Asp-Phe-Ile- LDFI) were shown to contribute to leptin binding site I and their mutations in alanine resulted in muteins acting as typical antagonists. We synthesized a small peptide based on the wild-type sequence of leptin binding site I (LDFI) and evaluated its efficacy in antagonizing leptin actions in breast cancer using in vitro and in vivo experimental models. The peptide LDFI abolished the leptin-induced anchorage-dependent and -independent growth as well as the migration of ERα-positive (MCF-7) and -negative (SKBR3) breast cancer cells. These results were well correlated with a reduction in the phosphorylation levels of leptin downstream effectors, as JAK2/STAT3/AKT/MAPK. Importantly, the peptide LDFI reversed the leptin-mediated up-regulation of its gene expression, as an additional mechanism able to enhance the peptide antagonistic activity. The described effects were specific for leptin signalling, since the developed peptide was not able to antagonize the other growth factors' actions on signalling activation, proliferation and migration. Finally, we showed that the LDFI pegylated peptide markedly reduced breast tumour growth in xenograft models. The unmodified peptide LDFI acting as a full leptin antagonist could become an attractive option for breast cancer treatment, especially in obese women.


Assuntos
Neoplasias da Mama/prevenção & controle , Proliferação de Células/efeitos dos fármacos , Leptina/antagonistas & inibidores , Oligopeptídeos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Sequência de Aminoácidos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Immunoblotting , Leptina/genética , Leptina/metabolismo , Células MCF-7 , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Oligopeptídeos/química , Fosforilação/efeitos dos fármacos , Polietilenoglicóis/química , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores para Leptina/antagonistas & inibidores , Receptores para Leptina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Carga Tumoral/efeitos dos fármacos
19.
Biochim Biophys Acta ; 1831(6): 1027-36, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23370576

RESUMO

The citrate carrier (CIC), a nuclear-encoded protein located in the mitochondrial inner membrane, plays an important metabolic role in the transport of acetyl-CoA from the mitochondrion to the cytosol in the form of citrate for fatty acid and cholesterol synthesis. Citrate has been reported to be essential for fibroblast differentiation into fat cells. Because peroxisome proliferator-activated receptor-gamma (PPARγ) is known to be one of the master regulators of adipogenesis, we aimed to study the regulation of CIC by the PPARγ ligand rosiglitazone (BRL) in 3T3-L1 fibroblasts and in adipocytes. We demonstrated that BRL up-regulated CIC mRNA and protein levels in fibroblasts, while it did not elicit any effects in mature adipocytes. The enhancement of CIC levels upon BRL treatment was reversed using the PPARγ antagonist GW9662, addressing how this effect was mediated by PPARγ. Functional experiments using a reporter gene containing rat CIC promoter showed that BRL enhanced CIC promoter activity. Mutagenesis studies, electrophoretic-mobility-shift assay and chromatin-immunoprecipitation analysis revealed that upon BRL treatment, PPARγ and Sp1 are recruited on the Sp1-containing region within the CIC promoter, leading to an increase in CIC expression. In addition, mithramycin, a specific inhibitor for Sp1-DNA binding activity, abolished the PPARγ-mediated up-regulation of CIC in fibroblasts. The stimulatory effects of BRL disappeared in mature adipocytes in which PPARγ/Sp1 complex recruited SMRT corepressor to the Sp1 site of the CIC promoter. Taken together, our results contribute to clarify the molecular mechanisms by which PPARγ regulates CIC expression during the differentiation stages of fibroblasts into mature adipocytes.


Assuntos
Adipócitos/metabolismo , Adipogenia/fisiologia , Fibroblastos/metabolismo , Mitocôndrias/metabolismo , PPAR gama/metabolismo , Proteínas Repressoras/genética , Células 3T3-L1 , Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Luciferases/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Correpressor 2 de Receptor Nuclear/antagonistas & inibidores , Correpressor 2 de Receptor Nuclear/genética , Correpressor 2 de Receptor Nuclear/metabolismo , PPAR gama/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rosiglitazona , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Tiazolidinedionas/farmacologia , Ativação Transcricional , Regulação para Cima
20.
Breast Cancer Res ; 16(1): R21, 2014 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-24552459

RESUMO

INTRODUCTION: The two isoforms of estrogen receptor (ER) alpha and beta play opposite roles in regulating proliferation and differentiation of breast cancers, with ER-alpha mediating mitogenic effects and ER-beta acting as a tumor suppressor. Emerging data have reported that androgen receptor (AR) activation inhibits ER-positive breast cancer progression mainly by antagonizing ER-alpha signaling. However, to date no studies have specifically evaluated a potential involvement of ER-beta in the inhibitory effects of androgens. METHODS: ER-beta expression was examined in human breast cancer cell lines using real-time PCR, Western blotting and small interfering RNA (siRNA) assays. Mutagenesis studies, electromobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) analysis were performed to assess the effects of mibolerone/AR on ER-beta promoter activity and binding. RESULTS: In this study, we demonstrate that mibolerone, a synthetic androgen ligand, up-regulates ER-beta mRNA and protein levels in ER-positive breast cancer cells. Transient transfection experiments, using a vector containing the human ER-beta promoter region, show that mibolerone increases basal ER-beta promoter activity. Site-directed mutagenesis and deletion analysis reveal that an androgen response element (ARE), TGTTCT motif located at positions -383 and -377, is critical for mibolerone-induced ER-beta up-regulation in breast cancer cells. This occurs through an increased recruitment of AR to the ARE site within the ER-beta promoter region, along with an enhanced occupancy of RNA polymerase II. Finally, silencing of ER-beta gene expression by RNA interference is able to partially reverse the effects of mibolerone on cell proliferation, p21 and cyclin D1 expression. CONCLUSIONS: Collectively, these data provide evidence for a novel mechanism by which activated AR, through an up-regulation of ER-beta gene expression, inhibits breast cancer cell growth.


Assuntos
Neoplasias da Mama/patologia , Receptor beta de Estrogênio/biossíntese , Receptores Androgênicos/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Ciclina D1/biossíntese , Ativação Enzimática , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Feminino , Humanos , Células MCF-7 , Mutagênese Sítio-Dirigida , Nandrolona/análogos & derivados , Nandrolona/farmacologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Ligação Proteica , Interferência de RNA , RNA Polimerase II/genética , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Congêneres da Testosterona/farmacologia , Regulação para Cima , Proteínas rho de Ligação ao GTP/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA