Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Regina Arantes-Rodrigues is active.

Publication


Featured researches published by Regina Arantes-Rodrigues.


Tumor Biology | 2014

Genomic characterization of three urinary bladder cancer cell lines: understanding genomic types of urinary bladder cancer.

Rosário Pinto-Leite; Isabel M. Carreira; Joana B. Melo; Susana Isabel Ferreira; Ilda Patrícia Ribeiro; Jaqueline Ferreira; Marco Filipe; Carina Bernardo; Regina Arantes-Rodrigues; Paula A. Oliveira; Lúcio Lara Santos

Several genomic regions are frequently altered and associated with the type, stage and progression of urinary bladder cancer (UBC). We present the characterization of 5637, T24 and HT1376 UBC cell lines by karyotyping, fluorescence in situ hybridization (FISH), array comparative genomic hybridization (aCGH) and multiplex ligation-dependent probe amplification (MLPA) analysis. Some cytogenetic anomalies present in UBC were found in the three cell lines, such as chromosome 20 aneuploidy and the loss of 9p21. Some gene loci losses (e.g. CDKN2A) and gains (e.g. HRAS, BCL2L1 and PTPN1) were coincident across all cell lines. Although some significant heterogeneity and complexity were detected between them, their genomic profiles exhibited a similar pattern to UBC. We suggest that 5637 and HT1376 represent the E2F3/RB1 pathway due to amplification of 6p22.3, concomitant with loss of one copy of RB1 and mutation of the remaining copy. The HT1376 presented a 10q deletion involving PTEN region and no alteration of PIK3CA region which, in combination with the inactivation of TP53, bears more invasive and metastatic properties than 5637. The T24 belongs to the alternative pathway of FGFR3/CCND1 by presenting mutated HRAS and over-represented CCND1. These cell lines cover the more frequent subtypes of UBC and are reliable models that can be used, as a group, in preclinical studies.


Urologic Oncology-seminars and Original Investigations | 2013

In vivo and in vitro effects of RAD001 on bladder cancer

C. Vasconcelos-Nóbrega; Rosário Pinto-Leite; Regina Arantes-Rodrigues; Rita Ferreira; Paulo Brochado; Maria Luís Cardoso; Carlos Palmeira; Alexandre Salvador; Catarina I. Guedes-Teixeira; Aura Colaço; Luis F. Palomino; Carlos Lopes; Lúcio Lara Santos; Paula A. Oliveira

OBJECTIVE To evaluate the influence of Everolimus (RAD001) on chemically induced urothelial lesions in mice and its influence on in vitro human bladder cancer cell lines. METHODS ICR male mice were given N-butyl-N-(4-hydroxybutyl) nitrosamine in drinking water for a period of 12 weeks. Subsequently, RAD001 was administered via oral gavage, for 6 weeks. At the end of the experiment, all the animals were sacrificed and tumor development was determined by means of histopathologic evaluation; mammalian target of rapamycin (mTOR) expressivity was evaluated by immunohistochemistry. Three human bladder cancer cell lines (T24, HT1376, and 5637) were treated using a range of RAD001 concentrations. MTT assay, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), and flow cytometry were used to assess cell proliferation, apoptosis index, and cell cycle analysis, respectively. Immunoblotting analysis of 3 cell line extracts using mTOR and Akt antibodies was performed in order to study the expression of Akt and mTOR proteins and their phosphorylated forms. RESULTS The incidence of urothelial lesions in animals treated with RAD001 was similar to those animals not treated. RAD001 did not block T24 and HT1376 cell proliferation or induce apoptosis. A reduction in cell proliferation rate and therefore G0/G1 phase arrest, as well as a statistically significant induction of apoptosis (P = 0.001), was only observed in the 5637 cell line. CONCLUSION RAD001 seems not to have a significant effect on chemically induced murine bladder tumors. The effect of RAD001 on tumor proliferation and apoptosis was achieved only in superficial derived bladder cancer cell line, no effect was observed in invasive cell lines.


Biomedicine & Pharmacotherapy | 2013

Everolimus combined with cisplatin has a potential role in treatment of urothelial bladder cancer.

Rosário Pinto-Leite; Regina Arantes-Rodrigues; Carlos Palmeira; Bruno Colaço; Carlos Lopes; Aura Colaço; Céu Costa; Vítor Moreira da Silva; Paula A. Oliveira; Lúcio Lara Santos

Cisplatin (CDDP)-based chemotherapy is a commonly treatment for advanced urothelial carcinoma. However, episodes of cisplatin resistance have been referenced. Recently it has been reported that everolimus (RAD001) could have an important role to play in bladder-cancer treatment and that mTOR inhibitors may restore chemosensitivity in resistant tumours. The aim of this study was to assess RAD001 in vitro ability to enhance CDDP cytotoxicity in three human bladder-cancer cell lines. Over the course of 72h, the cells were exposed to different concentrations of CDDP and RAD001, isolated or combined. Treatment with CDDP statistically (P<0.05) decreased cell proliferation in cell lines in a dose-dependent manner. The anti-proliferative activity of CDDP used in combination with RAD001 was statistically significant (P<0.05) in the cell lines at all concentrations tested. RAD001 had a therapeutic effect when used in combination with CDDP and could therefore be a useful anti-cancer drug combination for patients with bladder cancer.


Journal of Toxicology and Environmental Health | 2012

Everolimus Enhances Gemcitabine-Induced Cytotoxicity in Bladder-Cancer Cell Lines

Rosário Pinto-Leite; Regina Arantes-Rodrigues; Carlos Palmeira; Isabel Gaivão; Maria Luís Cardoso; Aura Colaço; Lúcio Lara Santos; Paula A. Oliveira

The purpose of this study was to determine whether everolimus, a rapamycin derivative, might significantly enhance the cytotoxicity of gemcitabine, an antitumor drug, in two human bladder-cancer cell lines. Human bladder-cancer T24 and 5637 cells were incubated with gemcitabine and everolimus in a range of concentrations either alone or in combination for 72 h. Flow cytometry, comet assay, MTT method and optical microscopy were used to assess cell proliferation, cell cycle, DNA damage, and morphological alterations. Gemcitabine exerted an inhibitory effect on T24 and 5637 cell proliferation, in a concentration-dependent manner. Everolimus significantly reduced proliferation of 5637 bladder cancer cells (IC30 at 1 μM), whereas T24 demonstrated marked resistance to everolimus treatment. A significant antiproliferative effect was obtained combining gemcitabine (100 nM) with everolimus (0.05–2 μM) with an arrest of cell cycle at S phase. Furthermore, an increase in frequency of DNA damage, apoptotic bodies, and apoptotic cells was observed when T24 and 5637 cancer cells were treated simultaneously with both drugs. Data show that in vitro combination produced a more potent antiproliferative effect when compared with single drugs.


Lab Animal | 2012

The effects of repeated oral gavage on the health of male CD-1 mice

Regina Arantes-Rodrigues; Andreia Henriques; Rosário Pinto-Leite; Ana I. Faustino-Rocha; Jacinta Pinho-Oliveira; C.I. Teixeira-Guedes; F. Seixas; A. Gama; Bruno Colaço; Aura Colaço; Paula A. Oliveira

Oral gavage is a widely used method for administering substances to animals in pharmacological and toxicological studies. The authors evaluated whether oral gavage causes behavioral indicators of stress, increased mortality rate, alterations in food and water consumption and body weight or histological lesions in CD-1 mice. Gavage was carried out once per d for 5 d per week over 6 consecutive weeks. The mortality rate of mice in this study was 15%. Mice subjected to gavage did not undergo changes in food or water consumption during the study, and their mean body weights and relative organ weights were similar to those of mice in the control group. Serum cortisol levels at the time of euthanasia in mice in both groups were within the normal range. Histopathology showed acute esophagitis and pleurisy, indicative of perforation of the esophagus, in the two mice that died but no abnormalities in the other mice. The results suggest that animal stress and mortality related to oral gavage can be minimized when the procedure is carried out by an experienced technician.


Food and Chemical Toxicology | 2011

High doses of olive leaf extract induce liver changes in mice

Regina Arantes-Rodrigues; Henriques A; Maria João Pires; Bruno Colaço; Ana M. Calado; P. Rema; Aura Colaço; T. Fernandes; P.L.F. De la Cruz; C. Lopes; Lio Fidalgo-Gonçalves; Sérgio M. F. Vilela; T. Pedrosa; Francisco Peixoto; Paula A. Oliveira

Virtually ever since it was first commercialized in 1995, there have been several studies focusing on the use of olive leaf extract (OLE) as a natural therapy and its medical properties. The aim of this study was to investigate the effects of three different concentrations of OLE on the function of mice livers over the course of 14 weeks. Female ICR mice were divided into four groups, depending on OLE concentration used: 0%, 0.25%, 0.5%, and 0.75%. Alanine aminotransferase, alkaline phosphatase, total bilirubin and albumin serum concentrations were all measured. Histopathological changes of the liver were observed after haematoxylin and eosin, reticulin, and Massons trichrome staining was carried out while liver mitochondrial bioenergetics were also evaluated. Alanine aminotransferase and alkaline phosphatase serum enzyme activities increased significantly in the groups in which 0.5% and 0.75% OLE concentrations were used. Histologically, all the groups exposed to OLE exhibited hyperplasia of the bile ducts, cholestasis, hepatocyte necrosis and inflammatory infiltrated. Hepatic fibrosis was observed in the groups featuring 0.5% and 0.75% OLE concentrations. The mitochondrial membrane potential, respiratory control ratio and ADP/O of samples from animals fed the higher OLE concentration was significantly decreased when compared to the control group.


Urologic Oncology-seminars and Original Investigations | 2014

Temsirolimus improves cytotoxic efficacy of cisplatin and gemcitabine against urinary bladder cancer cell lines

Rosário Pinto-Leite; Regina Arantes-Rodrigues; Rita Ferreira; Carlos M. Palmeira; Aura Colaço; Vítor Moreira da Silva; Paula A. Oliveira; Lúcio Lara Santos

OBJECTIVES To analyze the cytotoxic action of temsirolimus using 3 established human bladder cancer cell lines and to assess whether temsirolimus potentiates the anticancer activity of gemcitabine and cisplatin. METHODS Temsirolimus (500, 1,000, 2,000, and 4,000 nM), in isolation, and combined with gemcitabine (100 nM) and cisplatin (2.5 µg/ml), was given to 5637, T24, and HT1376 bladder cancer cell lines. Cell proliferation, autophagy, early apoptosis, and cell cycle distribution were analyzed after a 72-hour period. The expression of mammalian target of rapamycin baseline, Akt, and their phosphorylated forms, before and after treatment with temsirolimus, was evaluated by immunoblotting. RESULTS Temsirolimus slightly decreased the bladder cancer cell proliferation in all 3 cell lines. No significant differences in the expression of mammalian target of rapamycin, Akt, and their phosphorylated forms because of temsirolimus exposure were found in the 3 cell lines. As part of a combined regime along with gemcitabine, and especially with cisplatin, there was a more pronounced antiproliferative effect. This pattern of response was similar to the other parameters analyzed (increased autophagy and apoptosis). Also, in the combined regime, an enhanced cell cycle arrest in the G0/G1 phase was observed. The non-muscle invasive 5637 bladder cancer cell line was most sensitive to both combinations. CONCLUSIONS Temsirolimus makes a moderate contribution in terms of cell proliferation, apoptosis, and autophagy. However, it does potentiate the activity of gemcitabine and particularly cisplatin. Therefore, cisplatin- or gemcitabine-based chemotherapy regimen used in combination with temsirolimus to treat bladder cancer represents a novel and valuable treatment option, which should be tested for future studies in urinary bladder xenograft models.


International Journal of Experimental Pathology | 2014

Cytokeratin 7/19 expression in N-diethylnitrosamine-induced mouse hepatocellular lesions: implications for histogenesis

Nuno Paula Santos; Paula A. Oliveira; Regina Arantes-Rodrigues; Ana I. Faustino-Rocha; Aura Colaço; Carlos Lopes; Rui M. Gil da Costa

Hepatocellular carcinoma (HCC) is a common malignancy with poor clinical outcome, whose histogenesis is the subject of intense debate. Specifically, expression of cytokeratins (CKs) 7 and 19, associated with aggressive biological behaviour, is proposed to reflect a possible progenitor cell origin or tumour dedifferentiation towards a primitive phenotype. This work addresses that problem by studying CKs 7 and 19 expression in N‐diethylnitrosamine (DEN)‐induced mouse HCCs. ICR mice were divided into six DEN‐exposed and six matched control groups. Samples were taken from each group at consecutive time points. Hyperplastic foci (13 lesions) occurred at 29–40 weeks (groups 8, 10 and 12) with diffuse dysplastic areas (19 lesions) and with one hepatocellular adenoma (HCA) (at 29 weeks). HCCs (4 lesions) were observed 40 weeks after the first DEN administration (group 12). CKs 7 and 19 showed identical expression patterns and located to large, mature hepatocytes, isolated or in small clusters. Hyperplastic foci and the single HCA were consistently negative for both markers, while dysplastic areas and HCCs were positive. These results support the hypothesis that CKs 7 and 19 expression in hepatocellular malignancies results from a dedifferentiation process rather than from a possible progenitor cell origin.


Journal of Toxicology and Environmental Health | 2014

Effects of Naproxen on Cell Proliferation and Genotoxicity in MG-63 Osteosarcoma Cell Line

Isabel Correia; Regina Arantes-Rodrigues; Rosário Pinto-Leite; Isabel Gaivão

The purpose of this study was to determine the efficacy of naproxen, a nonsteroidal anti-inflammatory drug, on the MG-63 human osteosarcoma cell line. MG-63 cells were exposed to naproxen in a wide range of concentrations of 0.03, 0.05, 0.1, 0.42, 0.83, and 1.67 mg/ml for 72 h. The activity of naproxen was assessed by the following assays: cell morphology; 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method; terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay; comet assay; and acridine orange and monodansylcadaverine (MDC) staining. Naproxen exerted a significant inhibitory effect on MG-63 cell proliferation, in a concentration-dependent manner, in all treatment groups compared with untreated cells. An increase in frequency of DNA damage, apoptotic bodies, apoptotic cells, and autophagic vacuoles was observed in MG-63-treated cells. Although future studies are needed, these findings suggest that naproxen may lead to improvements in treatment of patients with osteosarcoma.


BioMed Research International | 2013

Synergistic Effect between Cisplatin and Sunitinib Malate on Human Urinary Bladder-Cancer Cell Lines

Regina Arantes-Rodrigues; Rosário Pinto-Leite; Lio Fidalgo-Gonçalves; Carlos M. Palmeira; Lúcio Lara Santos; Aura Colaço; Paula A. Oliveira

The aim of this paper is to analyse sunitinib malate in vitro ability to enhance cisplatin cytotoxicity in T24, 5637, and HT1376 human urinary bladder-cancer cell lines. Cells were treated with cisplatin (3, 6, 13, and 18 μM) and sunitinib malate (1, 2, 4, 6, and 20 μM), either in isolation or combined, over the course of 72 hours. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay, acridine orange, and monodansylcadaverine staining and flow cytometry were performed. The combination index (CI) was calculated based on the Chou and Talalay method. In isolation, cisplatin and sunitinib malate statistically (P < 0.05) decrease cell viability in all cell lines in a dose-dependent manner, with the presence of autophagic vacuoles. A cell cycle arrest in early S-phase and in G0/G1-phase was also found after exposure to cisplatin and sunitinib malate, in isolation, respectively. Treatment of urinary bladder-cancer cells with a combination of cisplatin and sunitinib malate showed a synergistic effect (CI < 1). Autophagy and apoptosis studies showed a greater incidence when the combined treatment was put into use. This hints at the possibility of a new combined therapeutic approach. If confirmed in vivo, this conjugation may provide a means of new perspectives in muscle-invasive urinary bladder cancer treatment.

Collaboration


Dive into the Regina Arantes-Rodrigues's collaboration.

Top Co-Authors

Avatar

Paula A. Oliveira

University of Trás-os-Montes and Alto Douro

View shared research outputs
Top Co-Authors

Avatar

Aura Colaço

University of Trás-os-Montes and Alto Douro

View shared research outputs
Top Co-Authors

Avatar

Lúcio Lara Santos

Instituto Português de Oncologia Francisco Gentil

View shared research outputs
Top Co-Authors

Avatar

Carlos Lopes

University of the Algarve

View shared research outputs
Top Co-Authors

Avatar

C. Vasconcelos-Nóbrega

Polytechnic Institute of Viseu

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ana I. Faustino-Rocha

University of Trás-os-Montes and Alto Douro

View shared research outputs
Top Co-Authors

Avatar

Bruno Colaço

University of Trás-os-Montes and Alto Douro

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge