Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Eur J Med Chem ; 188: 112030, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31945643

RESUMEN

Ruthenium-based complexes currently attract great attention as they hold promise to replace platinum-based drugs as a first line cancer treatment. Whereas ruthenium arene complexes are some of the most studied species for their potential anticancer properties, other types of ruthenium complexes have been overlooked for this purpose. Here, we report the synthesis and characterization of Ru(II) cyclopentadienyl (Cp), Ru(II) cyclooctadienyl (COD) and Ru(III) complexes bearing anastrozole or letrozole ligands, third-generation aromatase inhibitors currently used for the treatment of estrogen receptor positive (ER +) breast cancer. Among these complexes, Ru(II)Cp 2 was the only one that displayed a high stability in DMSO and in cell culture media and consequently, the only complex for which the in vitro and in vivo biological activities were investigated. Unlike anastrozole alone, complex 2 was considerably cytotoxic in vitro (IC50 values < 1 µM) in human ER + breast cancer (T47D and MCF7), triple negative breast cancer (TNBC) (MBA-MB-231), and in adrenocortical carcinoma (H295R) cells. Theoretical (docking simulation) and experimental (aromatase catalytic activity) studies suggested that an interaction between 2 and the aromatase enzyme was not likely to occur and that the bulkiness of the PPh3 ligands could be an important factor preventing the complex to reach the active site of the enzyme. Exposure of zebrafish embryos to complex 2 at concentrations around its in vitro cytotoxicity IC50 value (0.1-1 µM) did not lead to noticeable signs of toxicity over 96 h, making it a suitable candidate for further in vivo investigations. This study confirms the potential of Ru(II)Cp complexes for breast cancer therapy, more specifically against TNBCs that are usually not responsive to currently used chemotherapeutic agents.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Complejos de Coordinación/farmacología , Ciclopentanos/farmacología , Rutenio/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Ciclopentanos/química , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Rutenio/química , Relación Estructura-Actividad , Células Tumorales Cultivadas , Pez Cebra/embriología
2.
Organometallics ; 38(3): 702-711, 2019 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-31762529

RESUMEN

Third-generation aromatase inhibitors such as anastrozole (ATZ) and letrozole (LTZ) are widely used to treat estrogen receptor-positive ER+ breast cancers in postmenopausal women. Investigating their ability to coordinate metals could lead to the emergence of a new category of anticancer drug candidates with a broader spectrum of pharmacological activities. In this study, a series of ruthenium (II) arene complexes bearing the aromatase inhibitor anastrozole was synthesized and characterized. Among these complexes, [Ru(η 6 -C6H6)(PPh3)(η 1 -ATZ)Cl]BPh4 (3) was found to be the most stable in cell culture media, to lead to the highest cellular uptake and in vitro cytotoxicity in two ER+ human breast cancer cell lines (MCF7 and T47D), and to induce a decrease in aromatase activity in H295R cells. Exposure of zebrafish embryos to complex 3 (12.5 µM) did not lead to noticeable signs of toxicity over 96 h, making it a suitable candidate for further in vivo investigations.

3.
J Steroid Biochem Mol Biol ; 195: 105470, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31509772

RESUMEN

Serotonin reuptake inhibitors (SRIs) are currently the main molecules prescribed to pregnant women that suffer from depression. Placental cells are exposed to SRIs via maternal blood, and we have previously shown that SRIs alter feto-placental steroidogenesis in an in vitro co-culture model. More specifically, serotonin (5-HT) regulates the estrogen biosynthetic enzyme aromatase (cytochrome P450 19; CYP19), which is disrupted by fluoxetine and its active metabolite norfluoxetine in BeWo choriocarcinoma cells. Based on molecular simulations, the present study illustrates that the SRIs fluoxetine, norfluoxetine, paroxetine, sertraline, citalopram and venlafaxine exhibit binding affinity for the active-site pocket of CYP19, suggesting potential competitive inhibition. Using BeWo cells and primary villous trophoblast cells isolated from normal term placentas, we compared the effects of the SRIs on CYP19 activity. We observed that paroxetine and sertraline induce aromatase activity in BeWo cells, while venlafaxine, fluoxetine, paroxetine and sertraline decrease aromatase activity in primary villous trophoblast. The effects of paroxetine and sertraline in primary villous trophoblasts were observed at the lower doses tested. We also showed that 5-HT and the 5-HT2A receptor agonist 2,5-dimethoxy-4-iodoamphetamine (DOI) induced CYP19 activity. An increase in phosphorylation of serine and tyrosine and a decrease in threonine phosphorylation of CYP19 was also associated with DOI treatment. Our results contribute to better understanding how 5-HT and SRIs interact with CYP19 and may affect estrogen production. Moreover, this study suggests that alteration of placental 5-HT levels due to depression and/or SRI treatment during pregnancy may be associated with disruption of placental estrogen production.


Asunto(s)
Aromatasa/metabolismo , Placenta/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina/farmacología , Células Cultivadas , Citalopram/farmacología , Femenino , Fluoxetina/análogos & derivados , Fluoxetina/farmacología , Humanos , Simulación del Acoplamiento Molecular , Paroxetina/farmacología , Placenta/metabolismo , Embarazo , Sertralina/farmacología , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Clorhidrato de Venlafaxina/farmacología
4.
Mol Cell Endocrinol ; 498: 110583, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31536780

RESUMEN

In this study, we determined whether estragole and its isomer trans-anethole interfered with feto-placental steroidogenesis in a human co-culture model composed of fetal-like adrenocortical (H295R) and placental trophoblast-like (BeWo) cells. Estragole and trans-anethole are considered the biologically active compounds within basil and fennel seed essential oils, respectively. After a 24 h exposure of the co-culture to 2.5, 5.2 and 25 µM estragole or trans-anethole, hormone concentrations of estradiol, estrone, dehydroepiandrosterone, androstenedione, progesterone and estriol were significantly increased. Using RT-qPCR, estragole and trans-anethole were shown to significantly alter the expression of several key steroidogenic enzymes, such as those involved in cholesterol transport and steroid hormone biosynthesis, including StAR, CYP11A1, HSD3B1/2, SULT2A1, and HSD17B1, -4, and -5. Furthermore, we provided mechanistic insight into the ability of estragole and trans-anethole to stimulate promoter-specific expression of CYP19 through activation of the PKA pathway in H295R cells and the PKC pathway in BeWo cells, in both cases associated with increased cAMP levels. Moreover, we show new evidence suggesting a role for progesterone in regulating steroid hormone biosynthesis through regulation of the StAR gene.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/metabolismo , Anisoles/farmacología , Feto/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Placenta/metabolismo , Esteroides/metabolismo , Neoplasias de la Corteza Suprarrenal/tratamiento farmacológico , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/tratamiento farmacológico , Carcinoma Corticosuprarrenal/metabolismo , Carcinoma Corticosuprarrenal/patología , Derivados de Alilbenceno , Aromatasa/genética , Aromatasa/metabolismo , Supervivencia Celular , Coriocarcinoma/tratamiento farmacológico , Coriocarcinoma/metabolismo , Coriocarcinoma/patología , Técnicas de Cocultivo , Estradiol Deshidrogenasas/genética , Estradiol Deshidrogenasas/metabolismo , Femenino , Feto/efectos de los fármacos , Aromatizantes/farmacología , Humanos , Aceites Volátiles/farmacología , Placenta/efectos de los fármacos , Embarazo , Células Tumorales Cultivadas
6.
Biochimie ; 161: 88-108, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30946949

RESUMEN

We have reviewed the scientific literature related to four diseases in which to serotonin (5-HT) is involved in the etiology, herein named 5-HT-linked diseases, and whose prevalence is influenced by estrogenic status: depression, migraine, irritable bowel syndrome and eating disorders. These diseases all have in common a sex-dimorphic prevalence, with women more frequently affected than men. The co-occurrence between these 5-HT-linked diseases suggests that they have common physiopathological mechanisms. In most 5-HT-linked diseases (except for anorexia nervosa and irritable bowel syndrome), a decrease in the serotonergic tone is observed and estrogens are thought to contribute to the improvement of symptoms by stimulating the serotonergic system. Human pregnancy is characterized by a unique 5-HT and estrogen synthesis by the placenta. Pregnancy-specific disorders, such as hyperemesis gravidarum, gestational diabetes mellitus and pre-eclampsia, are associated with a hyperserotonergic state and decreased estrogen levels. Fetal programming of 5-HT-linked diseases is a complex phenomenon that involves notably fetal-sex differences, which suggest the implication of sex steroids. From a mechanistic point of view, we hypothesize that estrogens regulate the serotonergic system, resulting in a protective effect against 5-HT-linked diseases, but that, in turn, 5-HT affects estrogen synthesis in an attempt to retrieve homeostasis. These two processes (5-HT and estrogen biosynthesis) are crucial for successful pregnancy outcomes, and thus, a disruption of this 5-HT-estrogen relationship may explain pregnancy-specific pathologies or pregnancy complications associated with 5-HT-linked diseases.


Asunto(s)
Estrógenos/metabolismo , Placenta/fisiopatología , Complicaciones del Embarazo/fisiopatología , Serotonina/metabolismo , Interacciones Farmacológicas , Femenino , Humanos , Placenta/metabolismo , Embarazo , Complicaciones del Embarazo/metabolismo
7.
Placenta ; 72-73: 62-73, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30501883

RESUMEN

INTRODUCTION: Between 2 and 10% of pregnant women are treated with selective serotonin-reuptake inhibitors (SSRIs) for depression. The extravillous trophoblasts (evTBs), which migrate and invade maternal tissues, are crucial for embryo implantation and remodeling of maternal spiral arteries. Poor migration/invasion of evTBs can cause serious pregnancy complications, yet the effects of SSRIs on these processes has never been studied. To determine the effects of five SSRIs (fluoxetine, norfluoxetine, citalopram, sertraline and venlafaxine) on migration/invasion, we used JEG-3 and HIPEC cells as evTB models. METHODS: Cells were treated with increasing concentrations (0.03-10 µM) of SSRIs. Cell proliferation was monitored using an impedance-based system and cell cycle by flow cytometry. Migration was determined using a scratch test, and metalloproteinase (MMP) activities, by zymography. Invasion markers were determined by RT-qPCR. RESULTS: Fluoxetine and sertraline (10 µM) significantly decreased cell proliferation by 94% and by 100%, respectively, in JEG-3 cells, and by 58.6% and 100%, respectively, in HIPEC cells. Norfluoxetine increased MMP-9 activity in JEG-3 cells by 2.0% at 0.03 µM and by 43.9% at 3 µM, but decreased MMP-9 activity in HIPEC cells by 63.7% at 3 µM. Sertraline at 0.03 µM increased mRNA level of TIMP-1 in JEG-3 cells by 36% and that of ADAM-10 by 85% and 115% at 0.3 and 3 µM, respectively. In HIPEC cells, venlafaxine at 0.03 and 0.3 µM, increased ADAM-10 mRNA levels by 156% and 167%, respectively. DISCUSSION: This study shows that SSRIs may affect evTBs homeostasis at therapeutic levels and provides guidance for future research.


Asunto(s)
Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Trofoblastos/efectos de los fármacos , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Coriocarcinoma , Femenino , Fluoxetina/efectos adversos , Fluoxetina/análogos & derivados , Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 9 de la Matriz/efectos de los fármacos , Metaloproteinasa 9 de la Matriz/metabolismo , Proteínas de la Membrana/genética , Modelos Biológicos , Embarazo , ARN Mensajero/análisis , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Sertralina/efectos adversos , Inhibidor Tisular de Metaloproteinasa-1/genética , Trofoblastos/fisiología , Clorhidrato de Venlafaxina/efectos adversos
8.
Invest New Drugs ; 36(4): 718-725, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29607466

RESUMEN

Prostate cancer is the second leading cause of cancer-related deaths in men in North America and there is an urgent need for development of more effective therapeutic treatments against this disease. We have recently shown that diindolylmethane (DIM) and several of its halogenated derivatives (ring-DIMs) induce death and protective autophagy in human prostate cancer cells. However, the in vivo efficacy of ring-DIMs and the use of autophagy inhibitors as adjuvant therapy have not yet been studied in vivo. The objective of this study was to determine these effects on tumor growth in nude CD-1 mice bearing bioluminescent androgen-independent PC-3 human prostate cancer cells. We found that chloroquine (CQ) significantly sensitized PC-3 cells to death in the presence of sub-toxic concentrations of DIM or 4,4'-Br2DIM in vitro. Moreover, a combination of DIM (10 mg/kg) and CQ (60 mg/kg), 3× per week, significantly decreased PC-3 tumor growth in vivo after 3 and 4 weeks of treatment. Furthermore, 4,4'-Br2DIM at 10 mg/kg (3× per week) significantly inhibited tumour growth after 4 weeks of treatment. Tissues microarray analysis showed that DIM alone or combined with CQ induced apoptosis marker TUNEL; the combination also significantly inhibited the cell proliferation marker Ki67. In conclusion, we have confirmed that DIM and 4,4'-Br2DIM are effective agents against prostate cancer in vivo and shown that inhibition of autophagy with CQ enhances the anticancer efficacy of DIM. Our results suggest that including selective autophagy inhibitors as adjuvants may improve the efficacy of existing and novel drug therapies against prostate cancer.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Brassicaceae/química , Indoles/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Verduras/química , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cloroquina/farmacología , Xenoinjertos/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Desnudos , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Toxicol Appl Pharmacol ; 349: 8-20, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29679653

RESUMEN

Selective serotonin-reuptake inhibitors (SSRIs) are the most commonly prescribed antidepressants during pregnancy. The human placenta is a highly specialized organ supporting normal growth and development of the fetus. Therefore, this study aims to analyze the effects of SSRIs on villous cytotrophoblasts cells, using BeWo cells and human placental trophoblast cells in primary culture. The SSRIs fluoxetine and its metabolite norfluoxetine, sertraline and venlafaxine did not affect BeWo cell proliferation and viability, nor the percentage of M30-positive (apoptotic) primary trophoblast cells. None of the SSRIs affected basal or forskolin-stimulated BeWo cell fusion, whereas sertraline and venlafaxine increased the fusion of primary villous trophoblasts. Sertraline and venlafaxine also modified human chorionic gonadotropin beta (ß-hCG) secretion by BeWo cells, whereas none of the SSRIs affected ß-hCG secretion in primary trophoblasts. Norfluoxetine increased CGB (chorionic gonadotropin beta) and GJA1 (gap junction protein alpha 1) levels of gene expression (biomarkers of syncytialization) in BeWo cells, whereas in primary trophoblasts none of the SSRIs tested affected the expression of these genes. This study shows that SSRIs affect villous trophoblast syncytialization in a structure- and concentration-dependent manner and suggests that certain SSRIs may compromise placental health. In addition, it highlights the importance of using primary trophoblast cells instead of "trophoblast -like" cell lines to assess the effects of medications on human villous trophoblast function.


Asunto(s)
Células Gigantes/efectos de los fármacos , Placenta/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Trofoblastos/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Fusión Celular , Línea Celular , Proliferación Celular/efectos de los fármacos , Gonadotropina Coriónica/metabolismo , Conexina 43/metabolismo , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Mitosis/efectos de los fármacos , Embarazo
10.
Environ Health Perspect ; 126(4): 047014, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29701941

RESUMEN

BACKGROUND: Aromatase (CYP19) is a key enzyme in estrogens biosynthesis. In the mammary gland, CYP19 gene is expressed at low levels under the regulation of its I.4 promoter. In hormone-dependent breast cancer, fibroblast cells surrounding the tumor express increased levels of CYP19 mRNA due to a decrease of I.4 promoter activity and an increase of PII, I.3, and I.7 promoter activity. Little is known about the effects of environmental chemicals on the promoter-specific CYP19 expression. OBJECTIVE: We aimed to determine the effects of two neonicotinoids (thiacloprid and imidacloprid) on promoter-specific CYP19 expression in Hs578t breast cancer cells and understand the signaling pathways involved. METHODS: Hs578t cells were exposed to various signaling pathway stimulants or neonicotinoids for 24 h. Promoter-specific expression of CYP19 was determined by real-time quantitative polymerase chain reaction and catalytic activity of aromatase by tritiated water release assay. RESULTS: To our knowledge, we are the first to demonstrate that the normal I.4 promoter and the breast cancer-relevant PII, I.3, and I.7 promoters of CYP19 are active in these cells. We found that the expression of CYP19 via promoters PII, I.3, and I.7 in Hs578t cells was, in part, dependent on the activation of two VEGF signaling pathways: mitogen-activated protein kinase (MAPK) 1/3 and phospholipase C (PLC). Exposure of Hs578t cells to environmental concentrations of imidacloprid and thiacloprid resulted in a switch in CYP19 promoter usage, involving inhibition of I.4 promoter activity and an increase of PII, I.3, and I.7 promoter-mediated CYP19 expression and aromatase catalytic activity. Greater effects were seen at lower concentrations. Our results suggest that thiacloprid and imidacloprid exert their effects at least partially by inducing the MAPK 1/3 and/or PLC pathways. CONCLUSIONS: We demonstrated in vitro that neonicotinoids may stimulate a change in CYP19 promoter usage similar to that observed in patients with hormone-dependent breast cancer. https://doi.org/10.1289/EHP2698.


Asunto(s)
Aromatasa/genética , Regulación Neoplásica de la Expresión Génica , Insecticidas/toxicidad , Neonicotinoides/toxicidad , Nitrocompuestos/toxicidad , Transducción de Señal/efectos de los fármacos , Tiazinas/toxicidad , Factor A de Crecimiento Endotelial Vascular/genética , Aromatasa/metabolismo , Línea Celular Tumoral , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
Biochimie ; 148: 12-17, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29474975

RESUMEN

Estrogen production by the human villous trophoblast is dependent on the biosynthetic enzyme aromatase (CYP19; CYP19A1) and is crucial for successful placental development and pregnancy outcome. Using villous cytotrophoblast cells (vCTs) freshly isolated from normal term placenta, we characterized the promoter-specific expression of CYP19A1 mRNA (derived from promoters I.1, I.4, I.8 or total transcript) and aromatase activity during villous trophoblast syncytialization. CYP19A1 mRNA levels and aromatase activity in vCTs reached a maximum after about 48 h of culture. The cAMP inducer forskolin (10 µM) and protein kinase C stimulant phorbol myristate acetate (1 µM) increased CYP19A1 mRNA levels by 1.8- and 1.6-fold, respectively, as well as inducing aromatase catalytic activity. Dexamethasone (100 nM) and vascular endothelial growth factor (5 ng/mL) decreased CYP19A1 mRNA levels, while having no effect on aromatase activity. Our results emphasize the importance of not solely studying CYP19A1 regulation and function at the mRNA level but also considering posttranslational mechanisms that alter the final catalytic activity of aromatase.


Asunto(s)
Aromatasa/genética , Aromatasa/metabolismo , Vellosidades Coriónicas/metabolismo , Regulación Enzimológica de la Expresión Génica , Trofoblastos/metabolismo , Diferenciación Celular , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Trofoblastos/citología
12.
Methods Mol Biol ; 1710: 219-231, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29197006

RESUMEN

The placenta is a key element during pregnancy for the health of the fetus and the mother, which justifies why placental studies are so important. One of the best models for placental studies is the primary cell culture of cytotrophoblast cells from human term placentas. In this chapter, we will detail firstly the isolation of cytotrophoblast cells, with tissue preparation, digestion, Percoll gradient, and cell freezing, and secondly the cell immunopurification and seeding.


Asunto(s)
Separación Celular/métodos , Placenta/citología , Trofoblastos/citología , Técnicas de Cultivo de Célula , Células Cultivadas , Criopreservación , Femenino , Humanos , Povidona/química , Embarazo , Dióxido de Silicio/química
13.
Methods Mol Biol ; 1710: 267-276, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29197009

RESUMEN

In vitro functional analyses of cells are widely used to investigate the molecular mechanisms involved in preeclampsia. Common cellular functions studied include adhesion, apoptosis, proliferation, migration, and invasion. At present, most researchers will use endpoint experimental assays that only allow the determination of cell function at a single time point, with the need to repeat the experiment for an alternate time point. Here, we describe an electrical impedance-based tool that allows real-time monitoring of cells, which enables the efficient assessment of multiple time points over the duration of a single experiment.


Asunto(s)
Técnicas Electroquímicas/instrumentación , Placenta/citología , Apoptosis , Adhesión Celular , Movimiento Celular , Proliferación Celular , Impedancia Eléctrica , Diseño de Equipo , Femenino , Humanos , Microelectrodos , Embarazo , Programas Informáticos
14.
Methods Mol Biol ; 1710: 295-304, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29197012

RESUMEN

Estrogens are produced in large amounts during pregnancy, as a result of a tightly regulated cooperation between the maternal and fetal adrenal cortex, which produce androgen precursors, and the placental villous trophoblast, which transforms these precursors into estrogens. These estrogens play an important role in proper placental function, in adaptation of the mother to pregnancy, as well as in adequate fetal development. Disruption of estrogen production is associated with poor pregnancy outcomes and fetal malformation or altered fetal programming. Pregnant women may be exposed to endocrine disruptors from environmental sources or medications, and it is crucial to study the effects of such compounds on feto-placental steroidogenesis. The H295R/BeWo co-culture model offers the opportunity to study these interactions, by making it possible to evaluate the effects of chemical exposures on androgen and estrogen biosynthesis, as well as on various other aspects of feto-placental communication.


Asunto(s)
Técnicas de Cocultivo/métodos , Estrógenos/metabolismo , Feto/metabolismo , Placenta/metabolismo , Trofoblastos/metabolismo , Carcinoma Corticosuprarrenal/metabolismo , Vías Biosintéticas , Línea Celular Tumoral , Coriocarcinoma/metabolismo , Femenino , Feto/citología , Humanos , Placenta/citología , Embarazo , Trofoblastos/citología
15.
Cell Signal ; 40: 172-182, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28923415

RESUMEN

3,3'-Diindolylmethane (DIM) and its synthetic halogenated derivatives 4,4'-Br2- and 7,7'-Cl2DIM (ring-DIMs) have recently been shown to induce protective autophagy in human prostate cancer cells. The mechanisms by which DIM and ring-DIMs induce autophagy have not been elucidated. As DIM is a mitochondrial ATP-synthase inhibitor, we hypothesized that DIM and ring-DIMs induce autophagy via alteration of intracellular AMP/ATP ratios and activation of AMP-activated protein kinase (AMPK) signaling in prostate cancer cells. We found that DIM and ring-DIMs induced autophagy was accompanied by increased autophagic vacuole formation and conversion of LC3BI to LC3BII in LNCaP and C42B human prostate cancer cells. DIM and ring-DIMs also induced AMPK, ULK-1 (unc-51-like autophagy activating kinase 1; Atg1) and acetyl-CoA carboxylase (ACC) phosphorylation in a time-dependent manner. DIM and the ring-DIMs time-dependently induced the oncogenic protein astrocyte-elevated gene 1 (AEG-1) in LNCaP and C42B cells. Downregulation of AEG-1 or AMPK inhibited DIM- and ring-DIM-induced autophagy. Pretreatment with ULK1 inhibitor MRT 67307 or siRNAs targeting either AEG-1 or AMPK potentiated the cytotoxicity of DIM and ring-DIMs. Interestingly, downregulation of AEG-1 induced senescence in cells treated with overtly cytotoxic concentrations of DIM or ring-DIMs and inhibited the onset of apoptosis in response to these compounds. In summary, we have identified a novel mechanism for DIM- and ring-DIM-induced protective autophagy, via induction of AEG-1 and subsequent activation of AMPK. Our findings could facilitate the development of novel drug therapies for prostate cancer that include selective autophagy inhibitors as adjuvants.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Moléculas de Adhesión Celular/genética , Indoles/administración & dosificación , Neoplasias de la Próstata/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Tumoral , Halogenación/efectos de los fármacos , Humanos , Indoles/química , Masculino , Proteínas de la Membrana , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
16.
Toxicol Appl Pharmacol ; 332: 15-24, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28750898

RESUMEN

Estrogen biosynthesis during pregnancy is dependent on the collaboration between the fetus producing the androgen precursors, and the placenta expressing the enzyme aromatase (CYP19). Disruption of estrogen production by contaminants may result in serious pregnancy outcomes. We used our recently developed in vitro co-culture model of fetoplacental steroidogenesis to screen the effects of three neonicotinoid insecticides on the catalytic activity of aromatase and the production of steroid hormones. A co-culture of H295R human adrenocortical carcinoma cells with fetal characteristics and BeWo human choriocarcinoma cells which display characteristics of the villous cytotrophoblast was exposed for 24h to various concentrations of three neonicotinoids: thiacloprid, thiamethoxam and imidacloprid. Aromatase catalytic activity was determined in both cell lines using the tritiated water-release assay. Hormone production was measured by ELISA. The three neonicotinoids induced aromatase activity in our fetoplacental co-culture and concordingly, estradiol and estrone production were increased. In contrast, estriol production was strongly inhibited by the neonicotinoids. All three pesticides induced the expression of CYP3A7 in H295R cells, and this induction was reversed by co-treatment of H295R cells with exogenous estriol. CYP3A7 is normally expressed in fetal liver and is a key enzyme involved in estriol synthesis. We suggest that neonicotinoids are metabolized by CYP3A7, thus impeding the 16α-hydroxylation of fetal DHEA(-sulfate), which is normally converted to estriol by placental aromatase. We successfully used the fetoplacental co-culture as a physiologically relevant tool to highlight the potential effects of neonicotinoids on estrogen production, aromatase activity and CYP3A7 expression during pregnancy.


Asunto(s)
Aromatasa/metabolismo , Técnicas de Cocultivo/métodos , Disruptores Endocrinos/toxicidad , Insecticidas/toxicidad , Placenta/efectos de los fármacos , Carcinoma Corticosuprarrenal , Línea Celular Tumoral , Coriocarcinoma/inducido químicamente , Coriocarcinoma/diagnóstico , Citocromo P-450 CYP3A/genética , Citocromo P-450 CYP3A/metabolismo , Estradiol/metabolismo , Estrógenos/metabolismo , Estrona/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Imidazoles/toxicidad , Neonicotinoides , Nitrocompuestos/toxicidad , Oxazinas/toxicidad , Placenta/metabolismo , Embarazo , Piridinas/toxicidad , Tiametoxam , Tiazinas/toxicidad , Tiazoles/toxicidad , Neoplasias Uterinas/inducido químicamente , Neoplasias Uterinas/diagnóstico
17.
Biochimie ; 140: 159-165, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28751217

RESUMEN

The role of placental serotonin has been an active topic of research notably because of its crucial role in brain development. However, which cell types synthesize serotonin in human placenta remains unknown. Moreover, it is not known if the two tryptophan hydroxylase isoforms (TPH1 and TPH2), the rate-limiting enzymes in serotonin biosynthesis, are expressed in placenta. Human placentas were obtained in first trimester or at term, and trophoblast cells were isolated and purified using a magnetic cell sorter and placed in primary culture. The tissue sublocalization of each TPH was determined by immunohistochemistry. TPH expression in primary villous trophoblasts was determined by PCR and immunoblotting, and serotonin secretion by LC-MS/MS. Villous cytotrophoblasts, syncytiotrophoblast, fetal capillaries, extravillous cytotrophoblasts, and decidual cells co-expressed TPH1 and TPH2. Moreover, mRNA and protein levels of both TPHs were detected in human primary trophoblast as well as in mouse placental tissues. Finally, human trophoblast cells were shown to produce serotonin de novo. This study demonstrates that both TPH1 and TPH2 are expressed in human and mouse placenta throughout pregnancy and helps to better understand the placental serotonin system, which is crucial for healthy pregnancy and fetal development. It is therefore important to further understand regulation of the placental serotonin system and how its disruption during pregnancy may impact the developing fetus and subsequent child programming.


Asunto(s)
Decidua/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Proteínas Gestacionales/biosíntesis , Trofoblastos/enzimología , Triptófano Hidroxilasa/biosíntesis , Animales , Decidua/citología , Femenino , Humanos , Isoenzimas/biosíntesis , Ratones , Embarazo , Serotonina/biosíntesis , Trofoblastos/citología
18.
Mol Cell Endocrinol ; 442: 32-39, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27890559

RESUMEN

The effects of fluoxetine, one of the most prescribed selective serotonin-reuptake inhibitors (SSRIs) during pregnancy, and its active metabolite norfluoxetine were studied on placental aromatase (CYP19) and feto-placental steroidogenesis. Fluoxetine did not alter estrogen secretion in co-culture of fetal-like adrenocortical (H295R) and trophoblast-like (BeWo) cells used as a model of the feto-placental unit, although it induced CYP19 activity, apparently mediated by the serotonin (5-HT)2A receptor/PKC signaling pathway. Norfluoxetine decreased estrogen secretion in the feto-placental co-culture and competitively inhibited catalytic CYP19 activity in BeWo cells. Decreased serotonin transporter (SERT) activity in the co-culture was comparable to 17ß-estradiol treatment of BeWo cells. This work shows that the complex interaction of fluoxetine and norfluoxetine with placental estrogen production, involves 5-HT-dependent and -independent mechanisms. Considering the crucial role of estrogens during pregnancy, our results raise concern about the impact of SSRI treatment on placental function and fetal health.


Asunto(s)
Estrógenos/metabolismo , Fluoxetina/análogos & derivados , Fluoxetina/farmacología , Placenta/efectos de los fármacos , Aromatasa/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo/métodos , Estradiol/metabolismo , Femenino , Humanos , Proteínas de Transporte de Membrana/metabolismo , Placenta/metabolismo , Embarazo , Proteína Quinasa C/metabolismo , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Transducción de Señal/efectos de los fármacos
19.
PeerJ ; 4: e2445, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27896021

RESUMEN

Lithocholic acid (LCA) is a secondary bile acid that is selectively toxic to human neuroblastoma, breast and prostate cancer cells, whilst sparing normal cells. We previously reported that LCA inhibited cell viability and proliferation and induced apoptosis and necrosis of androgen-dependent LNCaP and androgen-independent PC-3 human prostate cancer cells. In the present study, we investigated the roles of endoplasmic reticulum (ER) stress, autophagy and mitochondrial dysfunction in the toxicity of LCA in PC-3 and autophagy deficient, androgen-independent DU-145 cells. LCA induced ER stress-related proteins, such as CCAAT-enhancer-binding protein homologous protein (CHOP), and the phosphorylation of eukaryotic initiation factor 2-alpha (p-eIF2α) and c-Jun N-terminal kinases (p-JNK) in both cancer cell-types. The p53 upregulated modulator of apoptosis (PUMA) and B cell lymphoma-like protein 11 (BIM) levels were decreased at overtly toxic LCA concentrations, although PUMA levels increased at lower LCA concentrations in both cell lines. LCA induced autophagy-related conversion of microtubule-associated proteins 1A/1B light chain 3B (LC3BI-LC3BII), and autophagy-related protein ATG5 in PC-3 cells, but not in autophagy-deficient DU-145 cells. LCA (>10 µM) increased levels of reactive oxygen species (ROS) concentration-dependently in PC-3 cells, whereas ROS levels were not affected in DU-145 cells. Salubrinal, an inhibitor of eIF2α dephosphorylation and ER stress, reduced LCA-induced CHOP levels slightly in PC-3, but not DU-145 cells. Salubrinal pre-treatment increased the cytotoxicity of LCA in PC-3 and DU-145 cells and resulted in a statistically significant loss of cell viability at normally non-toxic concentrations of LCA. The late-stage autophagy inhibitor bafilomycin A1 exacerbated LCA toxicity at subtoxic LCA concentrations in PC-3 cells. The antioxidant α-tocotrienol strongly inhibited the toxicity of LCA in PC-3 cells, but not in DU-145 cells. Collectively, although LCA induces autophagy and ER stress in PC-3 cells, these processes appear to be initially of protective nature and subsequently consequential to, but not critical for the ROS-mediated mitochondrial dysfunction and cytotoxicity of LCA. The full mechanism of LCA-induced mitochondrial dysfunction and cytotoxicity in the similarly sensitive DU-145 cells remains to be elucidated.

20.
J Vis Exp ; (113)2016 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-27500522

RESUMEN

This protocol describes how villous cytotrophoblast cells are isolated from placentas at term by successive enzymatic digestions, followed by density centrifugation, media gradient isolation and immunomagnetic purification. As observed in vivo, mononucleated villous cytotrophoblast cells in primary culture differentiate into multinucleated syncytiotrophoblast cells after 72 hr. Compared to normoxia (8% O2), villous cytotrophoblast cells that undergo hypoxia/reoxygenation (0.5% / 8% O2) undergo increased oxidative stress and intrinsic apoptosis, similar to that observed in vivo in pregnancy complications such as preeclampsia, preterm birth, and intrauterine growth restriction. In this context, primary villous trophoblasts cultured under hypoxia/reoxygenation conditions represent a unique experimental system to better understand the mechanisms and signalling pathways that are altered in human placenta and facilitate the search for effective drugs that protect against certain pregnancy disorders. Human villous trophoblasts produce melatonin and express its synthesizing enzymes and receptors. Melatonin has been suggested as a treatment for preeclampsia and intrauterine growth restriction because of its protective antioxidant effects. In the primary villous cytotrophoblast cell model described in this paper, melatonin has no effect on trophoblast cells in normoxic state but restores the redox balance of syncytiotrophoblast cells disrupted by hypoxia/reoxygenation. Thus, human villous trophoblast cells in primary culture are an excellent approach to study the mechanisms behind the protective effects of melatonin on placental function during hypoxia/reoxygenation.


Asunto(s)
Trofoblastos , Apoptosis , Hipoxia de la Célula , Células Cultivadas , Femenino , Humanos , Melatonina , Placenta , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA