Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Publication year range
1.
Genome Med ; 8(1): 88, 2016 08 24.
Article in English | MEDLINE | ID: mdl-27553366

ABSTRACT

BACKGROUND: Cancer patients often show no or only modest benefit from a given therapy. This major problem in oncology is generally attributed to the lack of specific predictive biomarkers, yet a global measure of cancer cell activity may support a comprehensive mechanistic understanding of therapy efficacy. We reasoned that network analysis of omic data could help to achieve this goal. METHODS: A measure of "cancer network activity" (CNA) was implemented based on a previously defined network feature of communicability. The network nodes and edges corresponded to human proteins and experimentally identified interactions, respectively. The edges were weighted proportionally to the expression of the genes encoding for the corresponding proteins and relative to the number of direct interactors. The gene expression data corresponded to the basal conditions of 595 human cancer cell lines. Therapeutic responses corresponded to the impairment of cell viability measured by the half maximal inhibitory concentration (IC50) of 130 drugs approved or under clinical development. Gene ontology, signaling pathway, and transcription factor-binding annotations were taken from public repositories. Predicted synergies were assessed by determining the viability of four breast cancer cell lines and by applying two different analytical methods. RESULTS: The effects of drug classes were associated with CNAs formed by different cell lines. CNAs also differentiate target families and effector pathways. Proteins that occupy a central position in the network largely contribute to CNA. Known key cancer-associated biological processes, signaling pathways, and master regulators also contribute to CNA. Moreover, the major cancer drivers frequently mediate CNA and therapeutic differences. Cell-based assays centered on these differences and using uncorrelated drug effects reveals novel synergistic combinations for the treatment of breast cancer dependent on PI3K-mTOR signaling. CONCLUSIONS: Cancer therapeutic responses can be predicted on the basis of a systems-level analysis of molecular interactions and gene expression. Fundamental cancer processes, pathways, and drivers contribute to this feature, which can also be exploited to predict precise synergistic drug combinations.


Subject(s)
Antineoplastic Agents/pharmacology , Drugs, Investigational/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Neoplasm Proteins/genetics , Prescription Drugs/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Female , Gene Expression Profiling , Gene Ontology , Humans , Molecular Sequence Annotation , Mutation , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
2.
Dis Model Mech ; 8(9): 1081-91, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26112171

ABSTRACT

miR-146a is a microRNA whose transcript levels are induced in the heart upon activation of NF-κB, a transcription factor induced by pro-inflammatory molecules (such as TNF-α) that is strongly related to the pathogenesis of cardiac disorders. The main goal of this study consisted of studying new roles of miR-146a in cardiac pathological processes caused by the pro-inflammatory cytokine TNF-α. Our results demonstrate that miR-146a transcript levels were sharply increased in cardiac ventricular tissue of transgenic mice with specific overexpression of TNF-α in the heart, and also in a cardiomyocyte cell line of human origin (AC16) exposed to TNF-α. Among all the in silico predicted miR-146a target genes, Fos mRNA and protein levels notably decreased after TNF-α treatment or miR-146a overexpression. These changes correlated with a diminution in the DNA-binding activity of AP-1, the Fos-containing transcription factor complex. Interestingly, AP-1 inhibition was accompanied by a reduction in matrix metalloproteinase (MMP)-9 mRNA levels in human cardiac cells. The specific regulation of this MMP by miR-146a was further confirmed at the secretion and enzymatic activity levels, as well as after anti-miR-mediated miR-146a inhibition. The results reported here demonstrate that Fos is a direct target of miR-146a activity and that downregulation of the Fos-AP-1 pathway by miR-146a has the capacity to inhibit MMP-9 activity. Given that MMP-9 is an AP-1 target gene involved in cardiac remodeling, myocardial dysfunction and progression of heart failure, these findings suggest that miR-146a might be a new and promising therapeutic tool for treating cardiac disorders associated with enhanced inflammation in the heart.


Subject(s)
Gene Expression Regulation , MicroRNAs/physiology , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Animals, Newborn , Cell Differentiation , Cell Line , Humans , Immune System , Inflammation , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Transgenic , Rats , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
PLoS One ; 10(1): e0115352, 2015.
Article in English | MEDLINE | ID: mdl-25590583

ABSTRACT

A key element to delineate the biology of individual tumors is the regulation of apoptosis. In this work, we functionally characterize two breast cancer associated genes, the proteasome 26S subunit ATPase 3 interacting protein (PSMC3IP) and the epithelial-stromal interaction 1 (EPSTI1), to explore their potential apoptotic role in breast cancer. We first explore the existence of direct physical interactions with annotated BC-apoptotic genes. Based on the generated interaction network, we examine several apoptotic markers to determine the effect of PSMC3IP and EPSTI1 gene expression modulation in two different human breast cancer cell lines to suggest potential molecular mechanisms to unveil their role in the disease. Our results show that PSMC3IP and EPSTI1 are able to modulate the extrinsic apoptotic pathway in estrogen receptor positive and triple negative breast cancer cell lines, highlighting them as potential therapeutic targets.


Subject(s)
Apoptosis/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Trans-Activators/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans
4.
Clín. investig. arterioscler. (Ed. impr.) ; 26(6): 255-267, nov.-dic. 2014. ilus, tab
Article in Spanish | IBECS | ID: ibc-129674

ABSTRACT

Introducción El estrés del retículo endoplasmático (RE) se ha relacionado con distintas enfermedades cardiovasculares, como la arteriosclerosis y la hipertrofia e insuficiencia cardíacas. Este estrés del RE altera la señalización de la insulina, contribuyendo al desarrollo de la resistencia a la insulina y la diabetes. Diversos estudios han demostrado que PPARalfa inhibe el estrés del RE, por lo que el objetivo de este trabajo consistió en investigar si la activación de este receptor nuclear era capaz de prevenir el estrés del RE inducido por ácidos grasos saturados en células cardíacas, así como los mecanismos implicados. Métodos: Cardiomiocitos humanos AC16 fueron tratados con palmitato en presencia de diferentes activadores e inhibidores de AMPK y PPARalfa. Para los estudios in vivo, ratones macho fueron alimentados con una dieta rica en grasa (HFD). Posteriormente, se determinó la presencia de distintos marcadores de estrés del RE en células cardíacas por medio del análisis de la expresión génica y la acumulación proteica. Resultados: El palmitato y la dieta HFD indujeron el estrés del RE en células cardíacas, pues incrementaron diversos marcadores de este, como son la expresión génica de ATF3, BiP/GRP78 y CHOP, el splicing de XBP1 y la fosforilación de IRE-1α y eIF2alfa. El tratamiento con Wy-14,643, un agonista de PPARalfa, previno el incremento del estrés del RE inducido por palmitato por medio de la activación de la AMPK. Conclusión: Wy-14,643 podría ser útil para prevenir el estrés del RE y las enfermedades cardiovasculares asociadas en pacientes obesos, e incluso durante la cardiomiopatía diabética, por medio de la activación de AMPK


Introduction Endoplasmic reticulum (ER) stress has been linked to several cardiovascular diseases, such as atherosclerosis, heart failure and cardiac hypertrophy. ER stress impairs insulin signalling, thus contributing to the development of insulin resistance and diabetes. Since several studies have reported that PPARalfa may inhibit ER stress, the main aim of this study consisted in investigating whether activation of this nuclear receptor is able to prevent lipid-induced ER stress in cardiac cells, as well as studying the mechanisms involved. Methods: A cardiomyocyte cell line of human origin, AC16, was treated with palmitate in the presence or absence of several AMPK and PPARα pharmacological agonists and antagonists. For the in vivo studies, wild-type male mice were fed a standard diet, or a high-fat diet (HFD), for two months. At the end of the experiments, several ER stress markers were assessed in cardiac cells or in the mice hearts, using real-time RT-PCR and Western-blot analyses. Results: The results demonstrate that both palmitate and the HFD induced ER stress in cardiac cells, since they upregulated the expression (ATF3, BiP/GRP78 and CHOP), splicing (sXBP1), and phosphorylation (IRE-1α and eIF2α) of several ER stress markers. Interestingly, treatment with the PPARalfa agonist Wy-14,643 prevented an increase in the majority of these ER stress markers in human cardiac cells by means of AMPK activation. Conclusion: These data indicate that PPARα activation by Wy-14,643 might be useful to prevent the harmful effects of ER stress and associated cardiovascular diseases in obese patients, and even during diabetic cardiomyopathy, by enhancing AMPK activity


Subject(s)
Animals , Mice , Endoplasmic Reticulum Stress , Peroxisome Proliferator-Activated Receptors/pharmacokinetics , Myocytes, Cardiac , Atherosclerosis/physiopathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , /analysis , Protective Agents/pharmacokinetics
5.
Clin Investig Arterioscler ; 26(6): 255-67, 2014.
Article in Spanish | MEDLINE | ID: mdl-24709376

ABSTRACT

INTRODUCTION: Endoplasmic reticulum (ER) stress has been linked to several cardiovascular diseases, such as atherosclerosis, heart failure and cardiac hypertrophy. ER stress impairs insulin signalling, thus contributing to the development of insulin resistance and diabetes. Since several studies have reported that PPARα may inhibit ER stress, the main aim of this study consisted in investigating whether activation of this nuclear receptor is able to prevent lipid-induced ER stress in cardiac cells, as well as studying the mechanisms involved. METHODS: A cardiomyocyte cell line of human origin, AC16, was treated with palmitate in the presence or absence of several AMPK and PPARα pharmacological agonists and antagonists. For the in vivo studies, wild-type male mice were fed a standard diet, or a high-fat diet (HFD), for two months. At the end of the experiments, several ER stress markers were assessed in cardiac cells or in the mice hearts, using real-time RT-PCR and Western-blot analyses. RESULTS: The results demonstrate that both palmitate and the HFD induced ER stress in cardiac cells, since they upregulated the expression (ATF3, BiP/GRP78 and CHOP), splicing (sXBP1), and phosphorylation (IRE-1α and eIF2α) of several ER stress markers. Interestingly, treatment with the PPARα agonist Wy-14,643 prevented an increase in the majority of these ER stress markers in human cardiac cells by means of AMPK activation. CONCLUSION: These data indicate that PPARα activation by Wy-14,643 might be useful to prevent the harmful effects of ER stress and associated cardiovascular diseases in obese patients, and even during diabetic cardiomyopathy, by enhancing AMPK activity.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endoplasmic Reticulum Stress/physiology , Myocytes, Cardiac/pathology , PPAR alpha/metabolism , Animals , Blotting, Western , Cell Line , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , PPAR alpha/agonists , Palmitates/administration & dosage , Pyrimidines/pharmacology , Real-Time Polymerase Chain Reaction
6.
Int J Cardiol ; 174(1): 110-8, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24767130

ABSTRACT

BACKGROUND: Chronic endoplasmic reticulum (ER) stress contributes to the apoptotic cell death in the myocardium, thereby playing a critical role in the development of cardiomyopathy. ER stress has been reported to be induced after high-fat diet feeding in mice and also after saturated fatty acid treatment in vitro. Therefore, since several studies have shown that peroxisome proliferator-activated receptor (PPAR)ß/δ inhibits ER stress, the main goal of this study consisted in investigating whether activation of this nuclear receptor was able to prevent lipid-induced ER stress in cardiac cells. METHODS AND RESULTS: Wild-type and transgenic mice with reduced PPARß/δ expression were fed a standard diet or a high-fat diet for two months. For in vitro studies, a cardiomyocyte cell line of human origin, AC16, was treated with palmitate and the PPARß/δ agonist GW501516. Our results demonstrate that palmitate induced ER stress in AC16 cells, a fact which was prevented after PPARß/δ activation with GW501516. Interestingly, the effect of GW501516 on ER stress occurred in an AMPK-independent manner. The most striking result of this study is that GW501516 treatment also upregulated the protein levels of beclin 1 and LC3II, two well-known markers of autophagy. In accordance with this, feeding on a high-fat diet or suppression of PPARß/δ in knockout mice induced ER stress in the heart. Moreover, PPARß/δ knockout mice also displayed a reduction in autophagic markers. CONCLUSION: Our data indicate that PPARß/δ activation might be useful to prevent the harmful effects of ER stress induced by saturated fatty acids in the heart by inducing autophagy.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Myocytes, Cardiac/drug effects , PPAR delta/pharmacology , PPAR-beta/pharmacology , Palmitates/pharmacology , Animals , Cells, Cultured , Humans , Male , Mice , Mice, Knockout , Thiazoles/pharmacology
7.
Int J Cardiol ; 167(6): 2507-16, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-22748497

ABSTRACT

BACKGROUND: Resveratrol is a grape polyphenol that prevents cardiac hypertrophy and protects the heart from ischemic injury, metabolic dysregulation, and inflammatory processes in several murine models. METHODS AND RESULTS: The aim of this study was to investigate the effects of resveratrol on the inflammatory processes in human cardiac AC16 cells in order to gain a better understanding of its cardioprotective mechanisms in the human heart. Resveratrol induced the DNA-binding activity of the pro-inflammatory transcription factor NF-κB in AC16 cells, and exacerbated the increase caused by tumor necrosis factor-α (TNF-α). In accordance with this, resveratrol increased the expression of the pro-inflammatory genes ICAM-1 (intercellular adhesion molecule-1) and TNF-α. In contrast, resveratrol decreased the expression of pro-inflammatory genes IL-6 (interleukin-6) and MCP-1 (monocyte chemoattractant protein-1). Likewise, resveratrol also induced inflammation in rat neonatal cardiomyocytes, and in the heart of mice fed a standard chow diet supplemented with resveratrol (1g/kg diet) for four months. Western-blot analyses revealed that NF-κB p65 subunit levels were upregulated in an IκB-dependent manner in the nuclei of resveratrol-treated human cardiac cells. Finally, resveratrol activated the signal transducer and activator of transcription 3 (STAT3) signaling and induced the expression of its anti-apoptotic downstream effector Bcl-xL, both involved in the cardioprotective survival activating factor enhancement (SAFE) pathway. CONCLUSIONS: Resveratrol enhanced NF-κB activity in human and murine cardiac cells, in a process that coincided with the activation of STAT3 and anti-apoptotic downstream effectors. Therefore, activation of the SAFE pathway by resveratrol might be involved in the cardioprotective effects of this compound.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Stilbenes/pharmacology , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Humans , Male , Mice , Rats , Rats, Sprague-Dawley , Resveratrol
SELECTION OF CITATIONS
SEARCH DETAIL
...