Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 22(1): 450, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741146

RESUMEN

BACKGROUND: Estetrol (E4) is a natural estrogen produced by the fetal liver during pregnancy. Due to its favorable safety profile, E4 was recently approved as estrogenic component of a new combined oral contraceptive. E4 is a selective ligand of estrogen receptor (ER)α and ERß, but its binding to the G Protein-Coupled Estrogen Receptor (GPER) has not been described to date. Therefore, we aimed to explore E4 action in GPER-positive Triple-Negative Breast Cancer (TNBC) cells. METHODS: The potential interaction between E4 and GPER was investigated by molecular modeling and binding assays. The whole transcriptomic modulation triggered by E4 in TNBC cells via GPER was explored through high-throughput RNA sequencing analyses. Gene and protein expression evaluations as well as migration and invasion assays allowed us to explore the involvement of the GPER-mediated induction of the plasminogen activator inhibitor type 2 (SERPINB2) in the biological responses triggered by E4 in TNBC cells. Furthermore, bioinformatics analysis was aimed at recognizing the biological significance of SERPINB2 in ER-negative breast cancer patients. RESULTS: After the molecular characterization of the E4 binding capacity to GPER, RNA-seq analysis revealed that the plasminogen activator inhibitor type 2 (SERPINB2) is one of the most up-regulated genes by E4 in a GPER-dependent manner. Worthy, we demonstrated that the GPER-mediated increase of SERPINB2 is engaged in the anti-migratory and anti-invasive effects elicited by E4 in TNBC cells. In accordance with these findings, a correlation between SERPINB2 levels and a good clinical outcome was found in ER-negative breast cancer patients. CONCLUSIONS: Overall, our results provide new insights into the mechanisms through which E4 can halt migratory and invasive features of TNBC cells.


Asunto(s)
Movimiento Celular , Estetrol , Regulación Neoplásica de la Expresión Génica , Inhibidor 2 de Activador Plasminogénico , Receptores de Estrógenos , Receptores Acoplados a Proteínas G , Transducción de Señal , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Movimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Estrógenos/metabolismo , Estetrol/farmacología , Estetrol/metabolismo , Femenino , Inhibidor 2 de Activador Plasminogénico/metabolismo , Unión Proteica/efectos de los fármacos , Invasividad Neoplásica
2.
J Transl Med ; 22(1): 597, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38937754

RESUMEN

BACKGROUND: Over the last two decades, tumor-derived RNA expression signatures have been developed for the two most commonly diagnosed tumors worldwide, namely prostate and breast tumors, in order to improve both outcome prediction and treatment decision-making. In this context, molecular signatures gained by main components of the tumor microenvironment, such as cancer-associated fibroblasts (CAFs), have been explored as prognostic and therapeutic tools. Nevertheless, a deeper understanding of the significance of CAFs-related gene signatures in breast and prostate cancers still remains to be disclosed. METHODS: RNA sequencing technology (RNA-seq) was employed to profile and compare the transcriptome of CAFs isolated from patients affected by breast and prostate tumors. The differentially expressed genes (DEGs) characterizing breast and prostate CAFs were intersected with data from public datasets derived from bulk RNA-seq profiles of breast and prostate tumor patients. Pathway enrichment analyses allowed us to appreciate the biological significance of the DEGs. K-means clustering was applied to construct CAFs-related gene signatures specific for breast and prostate cancer and to stratify independent cohorts of patients into high and low gene expression clusters. Kaplan-Meier survival curves and log-rank tests were employed to predict differences in the outcome parameters of the clusters of patients. Decision-tree analysis was used to validate the clustering results and boosting calculations were then employed to improve the results obtained by the decision-tree algorithm. RESULTS: Data obtained in breast CAFs allowed us to assess a signature that includes 8 genes (ITGA11, THBS1, FN1, EMP1, ITGA2, FYN, SPP1, and EMP2) belonging to pro-metastatic signaling routes, such as the focal adhesion pathway. Survival analyses indicated that the cluster of breast cancer patients showing a high expression of the aforementioned genes displays worse clinical outcomes. Next, we identified a prostate CAFs-related signature that includes 11 genes (IL13RA2, GDF7, IL33, CXCL1, TNFRSF19, CXCL6, LIFR, CXCL5, IL7, TSLP, and TNFSF15) associated with immune responses. A low expression of these genes was predictive of poor survival rates in prostate cancer patients. The results obtained were significantly validated through a two-step approach, based on unsupervised (clustering) and supervised (classification) learning techniques, showing a high prediction accuracy (≥ 90%) in independent RNA-seq cohorts. CONCLUSION: We identified a huge heterogeneity in the transcriptional profile of CAFs derived from breast and prostate tumors. Of note, the two novel CAFs-related gene signatures might be considered as reliable prognostic indicators and valuable biomarkers for a better management of breast and prostate cancer patients.


Asunto(s)
Neoplasias de la Mama , Fibroblastos Asociados al Cáncer , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Próstata , Humanos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Masculino , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Pronóstico , Transcriptoma/genética , Perfilación de la Expresión Génica , Análisis por Conglomerados , Resultado del Tratamiento , Persona de Mediana Edad , Estimación de Kaplan-Meier
3.
Int J Mol Sci ; 25(1)2023 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-38203608

RESUMEN

Arylalkane-derived prodrugs of arylacetic acids are a small group of substances that have long been known for their anti-inflammatory action. Despite their ease of synthesis and good potential for the development of new potent and safe anti-inflammatory agents, this group of substances has not received much attention from researchers so far. Therefore, representative arylalkane derivatives were investigated through molecular docking techniques to verify the possible hepatic activation mode toward active metabolites by CYP1A2. In this regard, arylalkanoic acid prodrugs were docked with a crystallographic structure of human CYP1A2, in which the enzyme is co-crystallized with the selective competitive inhibitor α-naphthoflavone BHF. Of note, all the examined compounds proved capable of interacting with the enzyme active site in a manner similar to Nabumetone, thus confirming that a productive metabolic transformation is feasible. On the basis of these findings, it is possible to argue that subtle differences in the way CYP1A2 accommodates the ligands depend on the fine details of their molecular structures. Overall, these data suggest that compounds simply formed by an aromatic moiety bearing an appropriate alkane-derived chain could lead to innovative anti-inflammatory agents.


Asunto(s)
Citocromo P-450 CYP1A2 , Profármacos , Humanos , Simulación del Acoplamiento Molecular , Antiinflamatorios/farmacología , Nabumetona , Profármacos/farmacología , Radiofármacos
4.
J Transl Med ; 20(1): 263, 2022 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672854

RESUMEN

BACKGROUND: Metabolic disorders are associated with increased incidence, aggressive phenotype and poor outcome of breast cancer (BC) patients. For instance, hyperinsulinemia is an independent risk factor for BC and the insulin/insulin receptor (IR) axis is involved in BC growth and metastasis. Of note, the anti-diabetic metformin may be considered in comprehensive therapeutic approaches in BC on the basis of its antiproliferative effects obtained in diverse pre-clinical and clinical studies. METHODS: Bioinformatics analysis were performed using the information provided by The Invasive Breast Cancer Cohort of The Cancer Genome Atlas (TCGA) project. The naturally immortalized BC cell line, named BCAHC-1, as well as cancer-associated fibroblasts (CAFs) derived from BC patients were used as model systems. In order to identify further mechanisms that characterize the anticancer action of metformin in BC, we performed gene expression and promoter studies as well as western blotting experiments. Moreover, cell cycle analysis, colony and spheroid formation, actin cytoskeleton reorganization, cell migration and matrigel drops evasion assays were carried out to provide novel insights on the anticancer properties of metformin. RESULTS: We first assessed that elevated expression and activation of IR correlate with a worse prognostic outcome in estrogen receptor (ER)-positive BC. Thereafter, we established that metformin inhibits the insulin/IR-mediated activation of transduction pathways, gene changes and proliferative responses in BCAHC-1 cells. Then, we found that metformin interferes with the insulin-induced expression of the metastatic gene CXC chemokine receptor 4 (CXCR4), which we found to be associated with poor disease-free survival in BC patients exhibiting high levels of IR. Next, we ascertained that metformin prevents a motile phenotype of BCAHC-1 cells triggered by the paracrine liaison between tumor cells and CAFs upon insulin activated CXCL12/CXCR4 axis. CONCLUSIONS: Our findings provide novel mechanistic insights regarding the anti-proliferative and anti-migratory effects of metformin in both BC cells and important components of the tumor microenvironment like CAFs. Further investigations are warranted to corroborate the anticancer action of metformin on the tumor mass toward the assessment of more comprehensive strategies halting BC progression, in particular in patients exhibiting metabolic disorders and altered insulin/IR functions.


Asunto(s)
Neoplasias de la Mama , Metformina , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Femenino , Humanos , Insulina/farmacología , Insulina/uso terapéutico , Metformina/farmacología , Metformina/uso terapéutico , Receptores CXCR4/metabolismo , Transducción de Señal , Microambiente Tumoral
5.
Int J Mol Sci ; 23(11)2022 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-35682675

RESUMEN

Increasing levels of estrogens across gestation are partly responsible for the physiological adaptations of the maternal vasculature to pregnancy. The G protein-coupled estrogen receptor (GPER) mediates acute vasorelaxing effects in the uterine vasculature, which may contribute to the regulation of uteroplacental blood flow. The aim of this study was to investigate whether GPER expression and vasorelaxation may occur following pregnancy. Elucidation of the functional signalling involved was also investigated. Radial uterine and third-order mesenteric arteries were isolated from non-pregnant (NP) and pregnant rats (P). GPER mRNA levels were determined and-concentration-response curve to the GPER-specific agonist, G1 (10-10-10-6 M), was assessed in arteries pre-constricted with phenylephrine. In uterine arteries, GPER mRNA expression was significantly increased and vasorelaxation to G1 was significantly enhanced in P compared with NP rats. Meanwhile, in mesenteric arteries, there was a similar order of magnitude in NP and P rats. Inhibition of L-type calcium channels and extracellular signal-regulated kinases 1/2 significantly reduced vasorelaxation triggered by G1 in uterine arteries. Increased GPER expression and GPER-mediated vasorelaxation are associated with the advancement of gestation in uterine arteries. The modulation of GPER is exclusive to uterine arteries, thus suggesting a physiological contribution of GPER toward the regulation of uteroplacental blood flow during pregnancy.


Asunto(s)
Calcio/metabolismo , Receptores de Estrógenos , Arteria Uterina , Animales , Dilatación , Estrógenos/metabolismo , Femenino , Proteínas de Unión al GTP/metabolismo , Sistema de Señalización de MAP Quinasas , Embarazo , ARN Mensajero/metabolismo , Ratas , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Arteria Uterina/metabolismo , Vasodilatación
6.
Inorg Chem ; 60(18): 14221-14229, 2021 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-34472350

RESUMEN

Achieving a precise control of the final structure of metal-organic frameworks (MOFs) is necessary to obtain desired physical properties. Here, we describe how the use of a metalloligand design strategy and a judicious choice of ligands inspired from nature is a versatile approach to succeed in this challenging task. We report a new porous chiral MOF, with the formula Ca5II{CuII10[(S,S)-aspartamox]5}·160H2O (1), constructed from Cu2+ and Ca2+ ions and aspartic acid-decorated ligands, where biometal Cu2+ ions are bridged by the carboxylate groups of aspartic acid moieties. The structure of MOF 1 reveals an infinite network of basket-like cages, built by 10 crystallographically distinct Cu(II) metal ions and five aspartamox ligands acting as bricks of a tubular motif, composed of seven basket-like cages each. The pillared hepta-packed cages generate pseudo-rhombohedral nanosized channels of ca. 0.7 and 0.4 nm along the b and a crystallographic axes. This intricate porous 3D network is anionic and chiral, each cage displaying receptor properties toward three-nuclear [Ca3(µ-H2O)4(H2O)17]6+ entities. 1 represents the first example of an extended porous structure based on essential biometals Cu2+ and Ca2+ ions together with aspartic acid as amino acid. 1 shows good biocompatibility, making it a good candidate to be used as a drug carrier, and hydrolyzes in acid water. The hypothesis has been further supported by an adsorption experiment here reported, as a proof-of-principle study, using dopamine hydrochloride as a model drug to follow the encapsulation process. Results validate the potential ability of 1 to act as a drug carrier. Thus, these make this MOF one of the few examples of biocompatible and degradable porous solid carriers for eventual release of drugs in the stomach stimulated by gastric low pH.


Asunto(s)
Ácido Aspártico/química , Materiales Biocompatibles/química , Estructuras Metalorgánicas/química , Materiales Biocompatibles/síntesis química , Humanos , Estructuras Metalorgánicas/síntesis química , Modelos Moleculares , Estructura Molecular , Células Tumorales Cultivadas
7.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-33946884

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive breast tumor subtype characterized by poor clinical outcome. In recent years, numerous advancements have been made to better understand the biological landscape of TNBC, though appropriate targets still remain to be determined. In the present study, we have determined that the expression levels of FGF2 and S100A4 are higher in TNBC with respect to non-TNBC patients when analyzing "The Invasive Breast Cancer Cohort of The Cancer Genome Atlas" (TCGA) dataset. In addition, we have found that the gene expression of FGF2 is positively correlated with S100A4 in TNBC samples. Performing quantitative PCR, Western blot, CRISPR/Cas9 genome editing, promoter studies, immunofluorescence analysis, subcellular fractionation studies, and ChIP assays, we have also demonstrated that FGF2 induces in TNBC cells the upregulation and secretion of S100A4 via FGFR1, along with the ERK1/2-AKT-c-Rel transduction signaling. Using conditioned medium from TNBC cells stimulated with FGF2, we have also ascertained that the paracrine activation of the S100A4/RAGE pathway triggers angiogenic effects in vascular endothelial cells (HUVECs) and promotes the migration of cancer-associated fibroblasts (CAFs). Collectively, our data provide novel insights into the action of the FGF2/FGFR1 axis through S100A4 toward stimulatory effects elicited in TNBC cells.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteínas de Neoplasias/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Proteína de Unión al Calcio S100A4/fisiología , Transducción de Señal/fisiología , Neoplasias de la Mama Triple Negativas/fisiopatología , Antígenos de Neoplasias/fisiología , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Factor 2 de Crecimiento de Fibroblastos/farmacología , Fibroblastos/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteínas Quinasas Activadas por Mitógenos/fisiología , Neovascularización Patológica/fisiopatología , Comunicación Paracrina , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-rel/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Transducción de Señal/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Células Tumorales Cultivadas
8.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096835

RESUMEN

Breast cancer represents the most frequently diagnosed malignancy in women worldwide. Various therapeutics are currently used in order to halt the progression of breast tumor, even though certain side effects may limit the beneficial effects. In recent years, many efforts have been addressed to the usefulness of natural compounds as anticancer agents due to their low toxicity. Resveratrol, a stilbene found in grapes, berries, peanuts and soybeans, has raised a notable interest for its antioxidant, anti-inflammatory, and antitumor properties. Here, we report the design, the synthesis and the characterization of the anticancer activity of a small series of imino N-aryl-substituted compounds that are analogues of resveratrol. In particular, the most active compound, named 3, exhibited anti-tumor activity in diverse types of breast cancer cells through the inhibition of the human topoisomerase II and the induction of apoptotic cell death. Therefore, the abovementioned compound maybe considered as a promising agent in more comprehensive treatments of breast cancer.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Resveratrol/análogos & derivados , Antineoplásicos/síntesis química , Disponibilidad Biológica , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/química , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo II/química , Femenino , Células HEK293 , Humanos , Iminas/química , Simulación del Acoplamiento Molecular , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/química , Resveratrol/farmacología , Inhibidores de Topoisomerasa I/química , Inhibidores de Topoisomerasa I/farmacología
9.
Int J Mol Sci ; 19(12)2018 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-30513833

RESUMEN

Breast cancer is the most prevalent cancer found in women living in developed countries. Endocrine therapy is the mainstay of treatment for hormone-responsive breast tumors (about 70% of all breast cancers) and implies the use of selective estrogen receptor modulators and aromatase inhibitors. In contrast, triple-negative breast cancer (TNBC), a highly heterogeneous disease that may account for up to 24% of all newly diagnosed cases, is hormone-independent and characterized by a poor prognosis. As drug resistance is common in all breast cancer subtypes despite the different treatment modalities, novel therapies targeting signaling transduction pathways involved in the processes of breast carcinogenesis, tumor promotion and metastasis have been subject to accurate consideration. G protein-coupled receptors (GPCRs) are the largest family of cell-surface receptors involved in the development and progression of many tumors including breast cancer. Here we discuss data regarding GPCR-mediated signaling, pharmacological properties and biological outputs toward breast cancer tumorigenesis and metastasis. Furthermore, we address several drugs that have shown an unexpected opportunity to interfere with GPCR-based breast tumorigenic signals.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Femenino , Humanos , Modelos Biológicos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Transducción de Señal
10.
Int J Mol Sci ; 19(7)2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29996493

RESUMEN

The Notch signaling pathway acts in both physiological and pathological conditions, including embryonic development and tumorigenesis. In cancer progression, diverse mechanisms are involved in Notch-mediated biological responses, including angiogenesis and epithelial-mesenchymal-transition (EMT). During EMT, the activation of cellular programs facilitated by transcriptional repressors results in epithelial cells losing their differentiated features, like cell­cell adhesion and apical­basal polarity, whereas they gain motility. As it concerns cancer epithelial cells, EMT may be consequent to the evolution of genetic/epigenetic instability, or triggered by factors that can act within the tumor microenvironment. Following a description of the Notch signaling pathway and its major regulatory nodes, we focus on studies that have given insights into the functional interaction between Notch signaling and either hypoxia or estrogen in breast cancer cells, with a particular focus on EMT. Furthermore, we describe the role of hypoxia signaling in breast cancer cells and discuss recent evidence regarding a functional interaction between HIF-1α and GPER in both breast cancer cells and cancer-associated fibroblasts (CAFs). On the basis of these studies, we propose that a functional network between HIF-1α, GPER and Notch may integrate tumor microenvironmental cues to induce robust EMT in cancer cells. Further investigations are required in order to better understand how hypoxia and estrogen signaling may converge on Notch-mediated EMT within the context of the stroma and tumor cells interaction. However, the data discussed here may anticipate the potential benefits of further pharmacological strategies targeting breast cancer progression.


Asunto(s)
Neoplasias de la Mama/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Notch/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Receptor Cross-Talk , Transducción de Señal
11.
J Cell Physiol ; 232(7): 1640-1649, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27607345

RESUMEN

The use of Doxorubicin (Dox), a frontline drug for many cancers, is often complicated by dose-limiting cardiotoxicity in approximately 20% of patients. The G-protein estrogen receptor GPER/GPR30 mediates estrogen action as the cardioprotection under certain stressful conditions. For instance, GPER activation by the selective agonist G-1 reduced myocardial inflammation, improved immunosuppression, triggered pro-survival signaling cascades, improved myocardial mechanical performance, and reduced infarct size after ischemia/reperfusion (I/R) injury. Hence, we evaluated whether ligand-activated GPER may exert cardioprotection in male rats chronically treated with Dox. 1 week of G-1 (50 µg/kg/day) intraperitoneal administration mitigated Dox (3 mg/kg/day) adverse effects, as revealed by reduced TNF-α, IL-1ß, LDH, and ROS levels. Western blotting analysis of cardiac homogenates indicated that G-1 prevents the increase in p-c-jun, BAX, CTGF, iNOS, and COX2 expression induced by Dox. Moreover, the activation of GPER rescued the inhibitory action elicited by Dox on the expression of BCL2, pERK, and pAKT. TUNEL assay indicated that GPER activation may also attenuate the cardiomyocyte apoptosis upon Dox exposure. Using ex vivo Langendorff perfused heart technique, we also found an increased systolic recovery and a reduction of both infarct size and LDH levels in rats treated with G-1 in combination with Dox respect to animals treated with Dox alone. Accordingly, the beneficial effects induced by G-1 were abrogated in the presence of the GPER selective antagonist G15. These data suggest that GPER activation mitigates Dox-induced cardiotoxicity, thus proposing GPER as a novel pharmacological target to limit the detrimental cardiac effects of Dox treatment. J. Cell. Physiol. 232: 1640-1649, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Cardiotónicos/uso terapéutico , Cardiotoxicidad/tratamiento farmacológico , Doxorrubicina/efectos adversos , Quinolinas/uso terapéutico , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Biomarcadores/metabolismo , Presión Sanguínea/efectos de los fármacos , Cardiotónicos/farmacología , Cardiotoxicidad/sangre , Cardiotoxicidad/patología , Cardiotoxicidad/fisiopatología , Diástole/efectos de los fármacos , Pruebas de Función Cardíaca/efectos de los fármacos , Humanos , Inflamación/patología , Interleucina-1beta/sangre , L-Lactato Deshidrogenasa/sangre , Ligandos , Masculino , Isquemia Miocárdica/sangre , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Quinolinas/farmacología , Ratas Wistar , Especies Reactivas de Oxígeno/sangre , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Factor de Necrosis Tumoral alfa/sangre , Función Ventricular/efectos de los fármacos
12.
Breast Cancer Res ; 19(1): 129, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29212519

RESUMEN

BACKGROUND: The G protein estrogen receptor GPER/GPR30 mediates estrogen action in breast cancer cells as well as in breast cancer-associated fibroblasts (CAFs), which are key components of microenvironment driving tumor progression. GPER is a transcriptional target of hypoxia inducible factor 1 alpha (HIF-1α) and activates VEGF expression and angiogenesis in hypoxic breast tumor microenvironment. Furthermore, IGF1/IGF1R signaling, which has angiogenic effects, has been shown to activate GPER in breast cancer cells. METHODS: We analyzed gene expression data from published studies representing almost 5000 breast cancer patients to investigate whether GPER and IGF1 signaling establish an angiocrine gene signature in breast cancer patients. Next, we used GPER-positive but estrogen receptor (ER)-negative primary CAF cells derived from patient breast tumours and SKBR3 breast cancer cells to investigate the role of GPER in the regulation of VEGF expression and angiogenesis triggered by IGF1. We performed gene expression and promoter studies, western blotting and immunofluorescence analysis, gene silencing strategies and endothelial tube formation assays to evaluate the involvement of the HIF-1α/GPER/VEGF signaling in the biological responses to IGF1. RESULTS: We first determined that GPER is co-expressed with IGF1R and with the vessel marker CD34 in human breast tumors (n = 4972). Next, we determined that IGF1/IGF1R signaling engages the ERK1/2 and AKT transduction pathways to induce the expression of HIF-1α and its targets GPER and VEGF. We found that a functional cooperation between HIF-1α and GPER is essential for the transcriptional activation of VEGF induced by IGF1. Finally, using conditioned medium from CAFs and SKBR3 cells stimulated with IGF1, we established that HIF-1α and GPER are both required for VEGF-induced human vascular endothelial cell tube formation. CONCLUSIONS: These findings shed new light on the essential role played by GPER in IGF1/IGF1R signaling that induces breast tumor angiogenesis. Targeting the multifaceted interactions between cancer cells and tumor microenvironment involving both GPCRs and growth factor receptors has potential in future combination anticancer therapies.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral , Factor A de Crecimiento Endotelial Vascular/metabolismo , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Factor I del Crecimiento Similar a la Insulina/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Mol Carcinog ; 56(2): 580-593, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27341075

RESUMEN

Zinc (Zn) is an essential trace mineral that contributes to the regulation of several cellular functions; however, it may be also implicated in the progression of breast cancer through different mechanisms. It has been largely reported that the classical estrogen receptor (ER), as well as the G protein estrogen receptor (GPER, previously known as GPR30) can exert a main role in the development of breast tumors. In the present study, we demonstrate that zinc chloride (ZnCl2 ) involves GPER in the activation of insulin-like growth factor receptor I (IGF-IR)/epidermal growth factor receptor (EGFR)-mediated signaling, which in turn triggers downstream pathways like ERK and AKT in breast cancer cells, and main components of the tumor microenvironment namely cancer-associated fibroblasts (CAFs). Further corroborating these findings, ZnCl2 stimulates a functional crosstalk of GPER with IGF-IR and EGFR toward the transcription of diverse GPER target genes. Then, we show that GPER contributes to the stimulatory effects induced by ZnCl2 on cell-cycle progression, proliferation, and migration of breast cancer cells as well as migration of CAFs. Together, our data provide novel insights into the molecular mechanisms through which zinc may exert stimulatory effects in breast cancer cells and CAFs toward tumor progression. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Neoplasias de la Mama/metabolismo , Fibroblastos Asociados al Cáncer/patología , Cloruros/metabolismo , Receptores ErbB/metabolismo , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Somatomedina/metabolismo , Transducción de Señal , Compuestos de Zinc/metabolismo , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/patología , Fibroblastos Asociados al Cáncer/metabolismo , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Femenino , Humanos
14.
Chem Biodivers ; 14(7)2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28217981

RESUMEN

This article reports the first study of the chemical composition, and antifungal and antiproliferative properties of the volatile extracts obtained by hydrodistillation of the flower heads and leaves of the traditional Kurdish medicinal plant Pterocephalus nestorianus Nábelek, collected in the wild. A total of 55 constituents, 43 of the flower heads' oil (PFO) and 46 of the leaves' oil (PLO), respectively, were identified by GC/MS, constituting 99.68% and 99.04% of the two oils, respectively. The oils were obtained in 0.15% and 0.10% yields (w/w), respectively, on air-dried vegetable material. The prevalent constituents of the PFO were α-terpineol (2.41%), α-linalool (6.42%), 6,10,14-trimethylpentadecan-2-one (2.59%), myristic acid (24.65%), and lauric acid (50.44%), while the major components of PLO were (E)-hex-2-enal (2.26%), (E)-hex-2-en-1-ol (2.04), myristic acid (34.03%), and lauric acid (50.35%). The two oils showed significant inhibitory and fungicidal activities against the medically important fungi Candida albicans, Candida tropicalis, Microsporum canis, and Trichophyton mentagrophytes, with minimum inhibitory concentration ranging from 0.7 to 3.3 mg/ml and minimum fungicidal concentration varying from 1.4 to 6.6 mg/ml. The antiproliferative activity of the two oils was assayed against one normal and six human tumor cell lines. Both oils showed selective cytotoxic activity, with IC50 values ranging from 1.4 to 3.3 µg/ml.


Asunto(s)
Antifúngicos/aislamiento & purificación , Caprifoliaceae/química , Antifúngicos/química , Antifúngicos/farmacología , Antineoplásicos Fitogénicos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Flores/química , Hongos/efectos de los fármacos , Cromatografía de Gases y Espectrometría de Masas , Humanos , Concentración 50 Inhibidora , Pruebas de Sensibilidad Microbiana , Aceites Volátiles/química , Hojas de la Planta/química
15.
Int J Mol Sci ; 18(12)2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29240722

RESUMEN

G protein-coupled receptors (GPCRs) have been implicated in transmitting signals across the extra- and intra-cellular compartments, thus allowing environmental stimuli to elicit critical biological responses. As GPCRs can be activated by an extensive range of factors including hormones, neurotransmitters, phospholipids and other stimuli, their involvement in a plethora of physiological functions is not surprising. Aberrant GPCR signaling has been regarded as a major contributor to diverse pathologic conditions, such as inflammatory, cardiovascular and neoplastic diseases. In this regard, solid tumors have been demonstrated to activate an angiogenic program that relies on GPCR action to support cancer growth and metastatic dissemination. Therefore, the manipulation of aberrant GPCR signaling could represent a promising target in anticancer therapy. Here, we highlight the GPCR-mediated angiogenic function focusing on the molecular mechanisms and transduction effectors driving the patho-physiological vasculogenesis. Specifically, we describe evidence for the role of heptahelic receptors and associated G proteins in promoting angiogenic responses in pathologic conditions, especially tumor angiogenesis and progression. Likewise, we discuss opportunities to manipulate aberrant GPCR-mediated angiogenic signaling for therapeutic benefit using innovative GPCR-targeted and patient-tailored pharmacological strategies.


Asunto(s)
Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Receptores Acoplados a Proteínas G/fisiología , Transducción de Señal , Animales , Progresión de la Enfermedad , Humanos , Modelos Biológicos , Neoplasias/irrigación sanguínea , Neoplasias/metabolismo , Neoplasias/fisiopatología , Neovascularización Patológica/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
17.
Breast Cancer Res Treat ; 146(2): 273-85, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24928526

RESUMEN

Tamoxifen resistance is a major clinical challenge in breast cancer treatment. Aromatase inhibitors are effective in women who progressed or recurred on tamoxifen, suggesting a role of local estrogen production by aromatase in driving tamoxifen-resistant phenotype. However, the link between aromatase activity and tamoxifen resistance has not yet been reported. We investigated whether long-term tamoxifen exposure may affect aromatase activity and/or expression, which may then sustain tamoxifen-resistant breast cancer cell growth. We employed MCF-7 breast cancer cells, tamoxifen-resistant MCF-7 cells (MCF-7 TR1 and TR2), SKBR-3 breast cancer cells, cancer-associated fibroblasts (CAFs1 and CAFs2). We used tritiated-water release assay, realtime-RT-PCR, and immunoblotting analysis for evaluating aromatase activity and expression; anchorage-independent assays for growth; reporter-gene, electrophoretic-mobility-shift, and chromatin-immunoprecipitation assays for promoter activity studies. We demonstrated an increased aromatase activity and expression, which supports proliferation in tamoxifen-resistant breast cancer cells. This is mediated by the G-protein-coupled receptor GPR30/GPER, since knocking-down GPER expression or treatment with a GPER antagonist reversed the enhanced aromatase levels induced by long-term tamoxifen exposure. The molecular mechanism was investigated in ER-negative, GPER/aromatase-positive SKBR3 cells, in which tamoxifen acts as a GPER agonist. Tamoxifen treatment increased aromatase promoter activity through an enhanced recruitment of c-fos/c-jun complex to AP-1 responsive elements located within the promoter region. As tamoxifen via GPER induced aromatase expression also in CAFs, this pathway may be involved in promoting aggressive behavior of breast tumors in response to tamoxifen treatment. Blocking estrogen production and/or GPER signaling activation may represent a valid option to overcome tamoxifen-resistance in breast cancers.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Aromatasa/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Tamoxifeno/farmacología , Aromatasa/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Células MCF-7 , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional
18.
Bioorg Med Chem Lett ; 24(1): 136-40, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24332626

RESUMEN

The synthesis and characterization of some new titanocene-complexes, having a ethenyl-phenoxide or a benzyl group as substituents of the cyclopentadienyl rings, are reported. The synthesized compounds have been evaluated for their cytotoxic potential against two human breast cancer cell lines, that is: MCF7 and SkBr3. Most of these compounds have shown significant cytotoxic effects, compared to cisplatin, in MTT-based cell tests.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Compuestos Organometálicos/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Células MCF-7 , Estructura Molecular , Compuestos Organometálicos/síntesis química , Compuestos Organometálicos/química , Estereoisomerismo , Relación Estructura-Actividad
19.
J Exp Clin Cancer Res ; 43(1): 171, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38886784

RESUMEN

BACKGROUND: The cyclin D1-cyclin dependent kinases (CDK)4/6 inhibitor palbociclib in combination with endocrine therapy shows remarkable efficacy in the management of estrogen receptor (ER)-positive and HER2-negative advanced breast cancer (BC). Nevertheless, resistance to palbociclib frequently arises, highlighting the need to identify new targets toward more comprehensive therapeutic strategies in BC patients. METHODS: BC cell lines resistant to palbociclib were generated and used as a model system. Gene silencing techniques and overexpression experiments, real-time PCR, immunoblotting and chromatin immunoprecipitation studies as well as cell viability, colony and 3D spheroid formation assays served to evaluate the involvement of the G protein-coupled estrogen receptor (GPER) in the resistance to palbociclib in BC cells. Molecular docking simulations were also performed to investigate the potential interaction of palbociclib with GPER. Furthermore, BC cells co-cultured with cancer-associated fibroblasts (CAFs) isolated from mammary carcinoma, were used to investigate whether GPER signaling may contribute to functional cell interactions within the tumor microenvironment toward palbociclib resistance. Finally, by bioinformatics analyses and k-means clustering on clinical and expression data of large cohorts of BC patients, the clinical significance of novel mediators of palbociclib resistance was explored. RESULTS: Dissecting the molecular events that characterize ER-positive BC cells resistant to palbociclib, the down-regulation of ERα along with the up-regulation of GPER were found. To evaluate the molecular events involved in the up-regulation of GPER, we determined that the epidermal growth factor receptor (EGFR) interacts with the promoter region of GPER and stimulates its expression toward BC cells resistance to palbociclib treatment. Adding further cues to these data, we ascertained that palbociclib does induce pro-inflammatory transcriptional events via GPER signaling in CAFs. Of note, by performing co-culture assays we demonstrated that GPER contributes to the reduced sensitivity to palbociclib also facilitating the functional interaction between BC cells and main components of the tumor microenvironment named CAFs. CONCLUSIONS: Overall, our results provide novel insights on the molecular events through which GPER may contribute to palbociclib resistance in BC cells. Additional investigations are warranted in order to assess whether targeting the GPER-mediated interactions between BC cells and CAFs may be useful in more comprehensive therapeutic approaches of BC resistant to palbociclib.


Asunto(s)
Neoplasias de la Mama , Quinasa 4 Dependiente de la Ciclina , Resistencia a Antineoplásicos , Piperazinas , Piridinas , Receptores de Estrógenos , Humanos , Piridinas/farmacología , Piridinas/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Piperazinas/farmacología , Piperazinas/uso terapéutico , Femenino , Receptores de Estrógenos/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Línea Celular Tumoral , Receptores Acoplados a Proteínas G/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Microambiente Tumoral
20.
J Biol Chem ; 287(52): 43234-45, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23135268

RESUMEN

Activation of lipid metabolism is an early event in carcinogenesis and a central hallmark of many tumors. Fatty acid synthase (FASN) is a key lipogenic enzyme catalyzing the terminal steps in the de novo biogenesis of fatty acids. In cancer cells, FASN may act as a metabolic oncogene, given that it confers growth and survival advantages to these cells, whereas its inhibition effectively and selectively kills tumor cells. Hormones such as estrogens and growth factors contribute to the transcriptional regulation of FASN expression also through the activation of downstream signaling and a cross-talk among diverse transduction pathways. In this study, we demonstrate for the first time that 17ß-estradiol (E2) and the selective GPER ligand G-1 regulate FASN expression and activity through the GPER-mediated signaling, which involved the EGF receptor/ERK/c-Fos/AP1 transduction pathway, as ascertained by using specific pharmacological inhibitors, performing gene-silencing experiments and ChIP assays in breast SkBr3, colorectal LoVo, hepatocarcinoma HepG2 cancer cells, and breast cancer-associated fibroblasts. In addition, the proliferative effects induced by E2 and G-1 in these cells involved FASN as the inhibitor of its activity, named cerulenin, abolished the growth response to both ligands. Our data suggest that GPER may be included among the transduction mediators involved by estrogens in regulating FASN expression and activity in cancer cells and cancer-associated fibroblasts that strongly contribute to cancer progression.


Asunto(s)
Estradiol/farmacología , Estrógenos/farmacología , Acido Graso Sintasa Tipo I/biosíntesis , Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Cerulenina/farmacología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Acido Graso Sintasa Tipo I/antagonistas & inhibidores , Acido Graso Sintasa Tipo I/genética , Inhibidores de la Síntesis de Ácidos Grasos/farmacología , Femenino , Fibroblastos/patología , Células Hep G2 , Humanos , Neoplasias/genética , Neoplasias/patología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA