13 results on '"Fabiana Ourique"'
Search Results
2. A novel role of MNT as a negative regulator of REL and the NF-κB pathway
- Author
-
Judit Liaño-Pons, M. Carmen Lafita-Navarro, Lorena García-Gaipo, Carlota Colomer, Javier Rodríguez, Alex von Kriegsheim, Peter J. Hurlin, Fabiana Ourique, M. Dolores Delgado, Anna Bigas, M. Lluis Espinosa, and Javier León
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract MNT, a transcription factor of the MXD family, is an important modulator of the oncoprotein MYC. Both MNT and MYC are basic-helix–loop–helix proteins that heterodimerize with MAX in a mutually exclusive manner, and bind to E-boxes within regulatory regions of their target genes. While MYC generally activates transcription, MNT represses it. However, the molecular interactions involving MNT as a transcriptional regulator beyond the binding to MAX remain unexplored. Here we demonstrate a novel MAX-independent protein interaction between MNT and REL, the oncogenic member of the NF-κB family. REL participates in important biological processes and it is altered in a variety of tumors. REL is a transcription factor that remains inactive in the cytoplasm in an inhibitory complex with IκB and translocates to the nucleus when the NF-κB pathway is activated. In the present manuscript, we show that MNT knockdown triggers REL translocation into the nucleus and thus the activation of the NF-κB pathway. Meanwhile, MNT overexpression results in the repression of IκBα, a bona fide REL target. Both MNT and REL bind to the IκBα gene on the first exon, suggesting its regulation as an MNT–REL complex. Altogether our data indicate that MNT acts as a repressor of the NF-κB pathway by two mechanisms: (1) retention of REL in the cytoplasm by MNT interaction, and (2) MNT-driven repression of REL-target genes through an MNT–REL complex. These results widen our knowledge about MNT biological roles and reveal a novel connection between the MYC/MXD and NF-κB pathways, two of the most prominent pathways in cancer.
- Published
- 2021
- Full Text
- View/download PDF
3. Correction: A novel role of MNT as a negative regulator of REL and the NF-κB pathway
- Author
-
Judit Liaño-Pons, M. Carmen Lafita-Navarro, Lorena García-Gaipo, Carlota Colomer, Javier Rodríguez, Alex von Kriegsheim, Peter J. Hurlin, Fabiana Ourique, M. Dolores Delgado, Anna Bigas, Lluis Espinosa, and Javier León
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2021
- Full Text
- View/download PDF
4. TOTAL CAROTENOID CONTENT OF SHRIMP COMMERCIALIZED IN FLORIANOPOLIS/SC AND EVALUATION OF COLOR PREFERENCE FOR CONSUMERS
- Author
-
Jane PARISENTI, Camila Cristina da Silveira BRITO, Vera Lúcia Cardoso Garcia TRAMONTE, Luiz Henrique BEIRÃO, Caroline Camila MOREIRA, and Fabiana OURIQUE
- Subjects
Shrimp ,astaxanthin ,consumer’ preference ,Nutrition. Foods and food supply ,TX341-641 - Abstract
The aim of this work was to evaluate total carotenoids content in shrimp commercialized in Florianopolis, SC, Brazil and to analyze consumers’ preference regarding to shrimp color. Samples of frozen (7) and fresh (7) shrimp from cultivation farms were obtained from fi sh market and public market. Total carotenoid content was determined spectophotometrically at 470mm. Concentration was calculated using astaxanthin standard curve. Two samples presenting the highest and the lowest total carotenoids content were selected for sensorial analysis (30 consumers), and were analyzed in colorimeter in order to confi rm color differences visually observed. Total carotenoids levels for fresh and frozen shrimp were 0.44 ± 0.23mg/100g and 0.48 ± 0.24mg/100g, respectively. Regarding to consumers’ preference, 90% (p
- Published
- 2011
5. Litopenaeus vannamei muscle carotenoids versus astaxanthin: A comparison of antioxidant activity and in vitro protective effects against lipid peroxidation
- Author
-
da Silva, Fabiana Ourique, Tramonte, Vera L.C.G., Parisenti, Jane, Lima-Garcia, Juliana F., Maraschin, Marcelo, and da Silva, Edson L.
- Published
- 2015
- Full Text
- View/download PDF
6. IP-Se-06, a Selenylated Imidazo[1,2-a]pyridine, Modulates Intracellular Redox State and Causes Akt/mTOR/HIF-1α and MAPK Signaling Inhibition, Promoting Antiproliferative Effect and Apoptosis in Glioblastoma Cells
- Author
-
Daniela C. dos Santos, Jamal Rafique, Sumbal Saba, Valdelúcia M. A. S. Grinevicius, Danilo W. Filho, Ariane Zamoner, Antonio L. Braga, Rozangela C. Pedrosa, and Fabiana Ourique
- Subjects
Aging ,Article Subject ,Cell Biology ,General Medicine ,Biochemistry - Abstract
Glioblastoma multiforme (GBM) is a notably lethal brain tumor associated with high proliferation rate and therapeutic resistance, while currently effective treatment options are still lacking. Imidazo[1,2-a]pyridine derivatives and organoselenium compounds are largely used in medicinal chemistry and drug development. This study is aimed at further investigating the antitumor mechanism of IP-Se-06 (3-((2-methoxyphenyl)selanyl)-7-methyl-2-phenylimidazol[1,2-a]pyridine), a selenylated imidazo[1,2-a]pyridine derivative in glioblastoma cells. IP-Se-06 exhibited high cytotoxicity against A172 cells ( I C 50 = 1.8 μ M ) and selectivity for this glioblastoma cell. The IP-Se-06 compound has pharmacological properties verified in its ADMET profile, especially related to blood-brain barrier (BBB) permeability. At low concentration (1 μM), IP-Se-06 induced intracellular redox state modulation with depletion of TrxR and GSH levels as well as inhibition of NRF2 protein. IP-Se-06 also decreased mitochondrial membrane potential, induced cytochrome c release, and chromatin condensation. Furthermore, IP-Se-06 induced apoptosis by decreasing levels of Bcl-xL while increasing levels of γ-H2AX and p53 proteins. Treatment with IP-Se-06 induced cell cycle arrest and showed antiproliferative effect by inhibition of Akt/mTOR/HIF-1α and ERK 1/2 signaling pathways. In addition, IP-Se-06 displayed significant inhibition of p38 MAPK and p-p38, leading to inhibition of inflammasome complex proteins (NLRP3 and caspase-1) in glioblastoma cells. These collective findings demonstrated that IP-Se-06 is a bioactive molecule that can be considered a candidate for the development of a novel drug for glioblastoma treatment.
- Published
- 2022
- Full Text
- View/download PDF
7. Correction: A novel role of MNT as a negative regulator of REL and the NF-κB pathway
- Author
-
Lluis Espinosa, Alex von Kriegsheim, Anna Bigas, Javier Rodriguez, M. Dolores Delgado, Judit Liaño-Pons, Javier León, Peter J. Hurlin, M. Carmen Lafita-Navarro, Lorena García-Gaipo, Carlota Colomer, and Fabiana Ourique
- Subjects
Cancer Research ,chemistry.chemical_compound ,Cell growth ,chemistry ,Molecular biology ,Cancer research ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Correction ,NF-κB ,Cancer genetics ,RC254-282 ,Negative regulator - Abstract
MNT, a transcription factor of the MXD family, is an important modulator of the oncoprotein MYC. Both MNT and MYC are basic-helix-loop-helix proteins that heterodimerize with MAX in a mutually exclusive manner, and bind to E-boxes within regulatory regions of their target genes. While MYC generally activates transcription, MNT represses it. However, the molecular interactions involving MNT as a transcriptional regulator beyond the binding to MAX remain unexplored. Here we demonstrate a novel MAX-independent protein interaction between MNT and REL, the oncogenic member of the NF-κB family. REL participates in important biological processes and it is altered in a variety of tumors. REL is a transcription factor that remains inactive in the cytoplasm in an inhibitory complex with IκB and translocates to the nucleus when the NF-κB pathway is activated. In the present manuscript, we show that MNT knockdown triggers REL translocation into the nucleus and thus the activation of the NF-κB pathway. Meanwhile, MNT overexpression results in the repression of IκBα, a bona fide REL target. Both MNT and REL bind to the IκBα gene on the first exon, suggesting its regulation as an MNT-REL complex. Altogether our data indicate that MNT acts as a repressor of the NF-κB pathway by two mechanisms: (1) retention of REL in the cytoplasm by MNT interaction, and (2) MNT-driven repression of REL-target genes through an MNT-REL complex. These results widen our knowledge about MNT biological roles and reveal a novel connection between the MYC/MXD and NF-κB pathways, two of the most prominent pathways in cancer.
- Published
- 2021
8. Albendazole as a promising molecule for tumor control
- Author
-
Jeferson Correia, Valdelúcia M.A.S. Grinevicius, Maicon Roberto Kviecinski, Rozangela Curi Pedrosa, Fabiana Ourique, D. Wilhelm Filho, Eduardo Benedetti Parisotto, and Luiza Sheyla Evenni Porfirio Will Castro
- Subjects
0301 basic medicine ,Programmed cell death ,Cell Survival ,DNA damage ,Clinical Biochemistry ,Antineoplastic Agents ,Apoptosis ,DNA fragmentation ,Oxidative phosphorylation ,Biology ,medicine.disease_cause ,Albendazole ,Biochemistry ,Cell cycle arrest ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Animals ,Humans ,Viability assay ,Carcinoma, Ehrlich Tumor ,lcsh:QH301-705.5 ,Cell Proliferation ,Membrane Potential, Mitochondrial ,lcsh:R5-920 ,Organic Chemistry ,Drug Repositioning ,Glutathione ,Antitumor ,Xenograft Model Antitumor Assays ,Molecular biology ,030104 developmental biology ,chemistry ,lcsh:Biology (General) ,Oxidative stress ,030220 oncology & carcinogenesis ,MCF-7 Cells ,Reactive Oxygen Species ,lcsh:Medicine (General) ,Research Paper ,DNA Damage - Abstract
This work evaluated the antitumor effects of albendazole (ABZ) and its relationship with modulation of oxidative stress and induction of DNA damage. The present results showed that ABZ causes oxidative cleavage on calf-thymus DNA suggesting that this compound can break DNA. ABZ treatment decreased MCF-7 cell viability (EC50=44.9 for 24 h) and inhibited MCF-7 colony formation (~67.5% at 5 μM). Intracellular ROS levels increased with ABZ treatment (~123%). The antioxidant NAC is able to revert the cytotoxic effects, ROS generation and loss of mitochondrial membrane potential of MCF-7 cells treated with ABZ. Ehrlich carcinoma growth was inhibited (~32%) and survival time was elongated (~50%) in animals treated with ABZ. Oxidative biomarkers (TBARS and protein carbonyl levels) and activity of antioxidant enzymes (CAT, SOD and GR) increased, and reduced glutathione (GSH) was depleted in animals treated with ABZ, indicating an oxidative stress condition, leading to a DNA damage causing phosphorylation of histone H2A variant, H2AX, and triggering apoptosis signaling, which was confirmed by increasing Bax/Bcl-xL rate, p53 and Bax expression. We propose that ABZ induces oxidative stress promoting DNA fragmentation and triggering apoptosis and inducing cell death, making this drug a promising leader molecule for development of new antitumor drugs., Graphical abstract fx1, Highlights • The ABZ redox signaling pathway was examined in cancer inhibition. • The oxidative stress can explain the ABZ antitumor mechanisms of action. • The ABZ oxidative stress modulation can be used for cancer therapeutics development. • ABZ can be used as a molecule prototype in possible drug repositioning.
- Published
- 2016
9. The MYC antagonist MNT autoregulates its expression and supports proliferation in MAX deficient cells
- Author
-
M. Dolores Delgado, Fabiana Ourique, Andrea Quintanilla, Julia Aresti, M. Carmen Lafita-Navarro, Rosa M. Blanco, Gabriel Bretones, Robert N. Eisenman, Peter J. Hurlin, Ignacio Varela, Javier León, Judit Liaño-Pons, and Patrick A. Carroll
- Subjects
0303 health sciences ,Cell growth ,Cell ,Biology ,Molecular biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Cytoplasm ,Transcription (biology) ,030220 oncology & carcinogenesis ,medicine ,Gene silencing ,Gene ,Transcription factor ,Nucleus ,030304 developmental biology - Abstract
MNT is a transcription factor of the MXD family. MNT-MAX dimers down-regulate genes by binding to E-box sequences, which can also be bound by MYC-MAX to activate transcription. MNT has been described as a modulator of MYC activity but little is known about MNT regulation and whether MNT has MAX-independent functions. Using a MAX deficient cell line and siRNA-mediated silencing of MAX, we show that in the absence of MAX, the total MNT levels are elevated and that MNT localizes both in the cytoplasm and the nucleus. In contrast, MNT is predominantly nuclear when MAX is expressed. MNT is required for optimal cell proliferation even in the absence of MAX, being the first report of a MAX-independent function of MNT. Interestingly, MNT forms homodimers and autoregulates its expression by repressing its own promoter. The tight MNT regulation and its activity in absence of MAX suggest its importance on cell homeostasis.
- Published
- 2017
- Full Text
- View/download PDF
10. DNA Damage and Inhibition of Akt Pathway in MCF-7 Cells and Ehrlich Tumor in Mice Treated with 1,4-Naphthoquinones in Combination with Ascorbate
- Author
-
Rozangela Curi Pedrosa, Valdelúcia M.A.S. Grinevicius, Karina Bettega Felipe, Pedro Buc Calderon, João Francisco Gomes Correia, Jaime A. Valderrama, Julio Benites, Maicon Roberto Kviecinski, Mirelle Sifroni Farias, David Ríos, Luiza Sheyla Evenni Porfirio Will Castro, Fabiana Ourique, and UCL - SSS/LDRI - Louvain Drug Research Institute
- Subjects
Male ,Aging ,Article Subject ,DNA damage ,Cell Survival ,Poly ADP ribose polymerase ,Antineoplastic Agents ,Ascorbic Acid ,Biology ,Biochemistry ,Histones ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,Animals ,Humans ,lcsh:QH573-671 ,Carcinoma, Ehrlich Tumor ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,chemistry.chemical_classification ,Reactive oxygen species ,Mice, Inbred BALB C ,Cell growth ,lcsh:Cytology ,Cell Biology ,General Medicine ,Ascorbic acid ,Molecular biology ,chemistry ,Cancer cell ,MCF-7 Cells ,Reactive Oxygen Species ,Proto-Oncogene Proteins c-akt ,Juglone ,Research Article ,Naphthoquinones ,DNA Damage - Abstract
The aim of this study was to enhance the understanding of the antitumor mechanism of 1,4-naphthoquinones and ascorbate. Juglone, phenylaminonaphthoquinone-7, and 9 (Q7/Q9) were evaluated for effects on CT-DNA and DNA of cancer cells. Evaluations in MCF-7 cells are DNA damage, ROS levels, viability, and proliferation. Proteins from MCF-7 lysates were immunoblotted for verifying PARP integrity,γH2AX, and pAkt. Antitumor activity was measured in Ehrlich ascites carcinoma-bearing mice. The same markers of molecular toxicity were assessedin vivo. The naphthoquinones intercalate into CT-DNA and caused oxidative cleavage, which is increased in the presence of ascorbate. Treatments caused DNA damage and reduced viability and proliferation of MCF-7 cells. Effects were potentiated by ascorbate. No PARP cleavage was observed. Naphthoquinones, combined with ascorbate, caused phosphorylation of H2AX and inhibited pAkt. ROS were enhanced in MCF-7 cells, particularly by the juglone and Q7 plus ascorbate. Ehrlich carcinoma was inhibited by juglone, Q7, or Q9, but the potentiating effect of ascorbate was reproducedin vivoonly in the cases of juglone and Q7, which caused up to 60% inhibition of tumor and the largest extension of survival. Juglone and Q7 plus ascorbate caused enhanced ROS and DNA damage and inhibited pAkt also in Ehrlich carcinoma cells.
- Published
- 2015
11. Sodium orthovanadate associated with pharmacological doses of ascorbate causes an increased generation of ROS in tumor cells that inhibits proliferation and triggers apoptosis
- Author
-
Günther, Tˆnia Mara Fischer, Kviecinski, Maicon Roberto, Baron, Carla Cristine, Felipe, Karina Bettega, Farias, Mirelle Sifroni, da Silva, Fabiana Ourique, Bücker, Nádia Cristina Falcão, Pich, Claus Tröger, Ferreira, Eduardo Antonio, Filho, Danilo Wilhelm, Verrax, Julien, Calderon, Pedro Buc, and Pedrosa, Rozangela Curi
- Published
- 2013
- Full Text
- View/download PDF
12. Substituted 3-acyl-2-phenylamino-1,4-naphthoquinones intercalate into DNA and cause genotoxicity through the increased generation of reactive oxygen species culminating in cell death.
- Author
-
FARIAS, MIRELLE SIFRONI, PICH, CLAUS TRÖGER, KVIECINSKI, MAICON ROBERTO, FALCÃO BUCKER, NÁDIA CRISTINA, FELIPE, KARINA BETTEGA, DA SILVA, FABIANA OURIQUE, FISHER GÜNTHER, TÂNIA MARA, CORREIA, JOÃO FRANCISCO, RÍOS, DAVID, BENITES, JULIO, VALDERRAMA, JAIME A., CALDERON, PEDRO BUC, and PEDROSA, ROZANGELA CURI
- Subjects
REACTIVE oxygen species ,CANCER cells ,CELL-mediated cytotoxicity ,ANTINEOPLASTIC agents ,BREAST cancer ,CELL lines ,CELL death - Abstract
Naphthoquinones interact with biological systems by generating reactive oxygen species (ROS) that can damage cancer cells. The cytotoxicity and the antitumor activity of 3-acyl-2-phenylamino-1,4-naphthoquinones (DPB1-DPB9) were evaluated in the MCF7 human breast cancer cell line and in male Ehrlich tumor-bearing Balb/c mice. DPB4 was the most cytotoxic derivative against MCF7 cells (EC
50 15 μM) and DPB6 was the least cytotoxic one (EC50 56 μM). The 1,4-naphthoquinone derivatives were able to cause DNA damage and promote DNA fragmentation as shown by the plasmid DNA cleavage assay (FII form). In addition, 1,4-naphthoquinone derivatives possibly interacted with DNA as intercalating agents, which was demonstrated by the changes caused in the fluorescence of the DNA-ethidium bromide complexes. Cell death of MCF7 cells induced by 3-acyl-2-phenylamino-1,4-naphthoquinones was mostly due to apoptosis. The DNA fragmentation and subsequent apoptosis may be correlated to the redox potential of the 1,4-naphthoquinone derivatives that, once present in the cell nucleus, led to the increased generation of ROS. Finally, certain 1,4-naphthoquinone derivatives and particularly DPB4 significantly inhibited the growth of Ehrlich ascites tumors in mice (73%). [ABSTRACT FROM AUTHOR]- Published
- 2014
- Full Text
- View/download PDF
13. Inhibition of tumor proliferation associated with cell cycle arrest caused by extract and fraction from Casearia sylvestris (Salicaceae).
- Author
-
Felipe, Karina Bettega, Kviecinski, Maicon Roberto, da Silva, Fabiana Ourique, Bücker, Nádia Falcão, Farias, Mirelle Sinfroni, Castro, Luiza Sheyla Evenni Porfirio Will, de Souza Grinevicius, Valdelúcia Maria Alves, Motta, Nadia Sandrini, Correia, João Francisco Gomes, Rossi, Maria Helena, and Pedrosa, Rozangela Curi
- Subjects
- *
MEDICINAL plants , *ALTERNATIVE medicine , *ANIMAL experimentation , *ANTINEOPLASTIC agents , *BIOLOGICAL assay , *BIOLOGICAL models , *BIOPHYSICS , *CHROMATOGRAPHIC analysis , *DOSE-effect relationship in pharmacology , *FLOW cytometry , *IMMUNOBLOTTING , *RESEARCH methodology , *MICE , *NUCLEAR magnetic resonance spectroscopy , *STAINS & staining (Microscopy) , *PLANT extracts , *DESCRIPTIVE statistics , *IN vitro studies , *PHARMACODYNAMICS ,BREAST tumor prevention - Abstract
Ethnopharmacological relevance Casearia sylvestris is a tree found in tropical America. In Brazil it is known mainly as Guaçatonga. Literature reports suggest that the leaves and other plant parts have been used by indigenous populations from South America in preparations, mainly aqueous or hydroethanolic macerations or decoctions, most times taken orally for the primary treatment of several diseases, including cancer. Aim of the study This article reports the results of an investigation about the antiproliferative effects of Casearia sylvestris on tumor cells in vitro and in vivo . Material and methods Aqueous ethanolic maceration and column chromatography were done to obtain a crude aqueous ethanolic extract (CAE) and a chloroform fraction (f-CHCl 3 ). The human breast cancer cell line MCF-7 was used in culture. In vitro , non-cytotoxic concentrations were determined by MTT assay and the antiproliferative effect was assessed by the colony forming unit assay using non-cytotoxic concentrations. Effects on the cell cycle were observed through flow cytometry using a propidium iodide kit. Casearin C was identified in f-CHCl 3 by chromatography and H 1 nuclear magnetic resonance. The effect on some key proteins of DNA damage (phosphorylation on the histone H2AX) and cell cycle control (p53, p16, cdk2) was evaluated through immunoblot. Antiproliferative effects in vivo were measured in tumor tissue from Ehrlich ascites-bearing mice through the 3 H-thymidine uptake assay and the trypan blue exclusion method. Results In vitro , EC 50 values found at 24 h on MCF-7 cells were 141 µg/mL for CAE and 66 µg/mL for f-CHCl 3 . Inhibition on proliferation was recorded at concentrations as low as 4 µg/mL in the case of the f-CHCl 3 (up to 40%) and up to 50% when CAE was added at 9 µg/mL. The cell cycle arrest was demonstrated by the reduction in terms of number of cells in phases G2/M and S, up to 38.9% and 51.9% when cells were treated with CAE, and 53.9% and 66.2%, respectively, when cells were treated with f-CHCl 3 . The number of cells in G1 was increased when the cells were treated with CAE (21.4%) or f-CHCl 3 (27.8%). Key proteins of cell cycle control were affected. The treatments caused activation of p53, p16 and DNA damage found by the appearance of bands corresponding to γ -H2AX. The treatments caused inhibition of cdk2. CAE and particularly f-CHCl 3 caused significant inhibition on tumor growth in mice (40% and 60%, respectively). Uptake of 3 H-thymidine, thus proliferation was reduced in tumor cells from mice treated with CAE (>30%) or f-CHCl 3 (up to 50%) compared to cells from control animals. Data from the trypan blue assay indicating a lower number of tumor cells in treated animals. From the overall, data from this study are in line with the traditional claims for the antitumor effect of Casearia sylvestris . Conclusions This investigation suggests that whether the extracts from Casearia sylvestris are cytotoxic at high concentrations, lower concentrations have antiproliferative effect and could be useful to complement conventional cytotoxic chemotherapy, and should be evaluated further. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.