Network


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

Hotspot


Dive into the research topics where Luciana Madeira da Silva is active.

Publication


Featured researches published by Luciana Madeira da Silva.


PLOS ONE | 2014

ALDH1A1 Maintains Ovarian Cancer Stem Cell-Like Properties by Altered Regulation of Cell Cycle Checkpoint and DNA Repair Network Signaling

Erhong Meng; Aparna Mitra; Kaushlendra Tripathi; Michael A. Finan; J.M. Scalici; Steve McClellan; Luciana Madeira da Silva; Eddie Reed; Lalita A. Shevde; Komaraiah Palle; Rodney P. Rocconi

Objective Aldehyde dehydrogenase (ALDH) expressing cells have been characterized as possessing stem cell-like properties. We evaluated ALDH+ ovarian cancer stem cell-like properties and their role in platinum resistance. Methods Isogenic ovarian cancer cell lines for platinum sensitivity (A2780) and platinum resistant (A2780/CP70) as well as ascites from ovarian cancer patients were analyzed for ALDH+ by flow cytometry to determine its association to platinum resistance, recurrence and survival. A stable shRNA knockdown model for ALDH1A1 was utilized to determine its effect on cancer stem cell-like properties, cell cycle checkpoints, and DNA repair mediators. Results ALDH status directly correlated to platinum resistance in primary ovarian cancer samples obtained from ascites. Patients with ALDHHIGH displayed significantly lower progression free survival than the patients with ALDHLOW cells (9 vs. 3 months, respectively p<0.01). ALDH1A1-knockdown significantly attenuated clonogenic potential, PARP-1 protein levels, and reversed inherent platinum resistance. ALDH1A1-knockdown resulted in dramatic decrease of KLF4 and p21 protein levels thereby leading to S and G2 phase accumulation of cells. Increases in S and G2 cells demonstrated increased expression of replication stress associated Fanconi Anemia DNA repair proteins (FANCD2, FANCJ) and replication checkpoint (pS317 Chk1) were affected. ALDH1A1-knockdown induced DNA damage, evidenced by robust induction of γ-H2AX and BAX mediated apoptosis, with significant increases in BRCA1 expression, suggesting ALDH1A1-dependent regulation of cell cycle checkpoints and DNA repair networks in ovarian cancer stem-like cells. Conclusion This data suggests that ovarian cancer cells expressing ALDH1A1 may maintain platinum resistance by altered regulation of cell cycle checkpoint and DNA repair network signaling.


Cancer Research | 2016

Immunoregulatory Protein B7-H3 Reprograms Glucose Metabolism in Cancer Cells by ROS-Mediated Stabilization of HIF1a

Sangbin Lim; Hao Liu; Luciana Madeira da Silva; Ritu Arora; Zixing Liu; Joshua B. Phillips; David C. Schmitt; Tung Vu; Steven McClellan; Yifeng Lin; Wensheng Lin; Gary A. Piazza; Øystein Fodstad; Ming Tan

B7-H3 is a member of B7 family of immunoregulatory transmembrane glycoproteins expressed by T cells. While B7-H3 overexpression is associated with poor outcomes in multiple cancers, it also has immune-independent roles outside T cells and its precise mechanistic contributions to cancer are unclear. In this study, we investigated the role of B7-H3 in metabolic reprogramming of cancer cells in vitro and in vivo We found that B7-H3 promoted the Warburg effect, evidenced by increased glucose uptake and lactate production in B7-H3-expressing cells. B7-H3 also increased the protein levels of HIF1α and its downstream targets, LDHA and PDK1, key enzymes in the glycolytic pathway. Furthermore, B7-H3 promoted reactive oxygen species-dependent stabilization of HIF1α by suppressing the activity of the stress-activated transcription factor Nrf2 and its target genes, including the antioxidants SOD1, SOD2, and PRX3. Metabolic imaging of human breast cancer xenografts in mice confirmed that B7-H3 enhanced tumor glucose uptake and tumor growth. Together, our results illuminate the critical immune-independent contributions of B7-H3 to cancer metabolism, presenting a radically new perspective on B7 family immunoregulatory proteins in malignant progression. Cancer Res; 76(8); 2231-42. ©2016 AACR.


ACS Medicinal Chemistry Letters | 2015

Synthesis of a New Peptide–Coumarin Conjugate:A Potential Agent against Cryptococcosis

Soraya Maria Zandim Maciel Dias Ferreira; Hellem Cristina Silva Carneiro; Hugo Vinícius de Andrade Lara; Rosemeire B. Alves; Jarbas M. Resende; Heloísa M. Oliveira; Luciana Madeira da Silva; Daniel Assis Santos; Rossimiriam Pereira de Freitas

Antimicrobial peptides (AMPs) are currently being investigated as potential sources of novel therapeutics against an increasing number of microorganisms resistant to conventional antibiotics. The conjugation of an AMP to other bioactive compounds is an interesting approach for the development of new derivatives with increased antimicrobial efficiency and broader spectra of action. In this work, the synthesis of a new peptide-coumarin conjugate via copper(I)-catalyzed azide-alkyne cycloaddition is described. The conjugate was assayed for in vitro cytotoxicity and displayed antifungal activity against Cryptococcus gattii and Cryptococcus neoformans. Additionally, the conjugate exhibited increased antifungal efficacy when compared with the individual peptide, coumarin, or triazole moieties. Treatment of C. gattii with the peptide-coumarin conjugate enhanced the production of reactive oxygen species, suggesting that the oxidative burst plays an important role in the mechanism of action of the conjugate.


PLOS ONE | 2014

21-Benzylidene Digoxin: A Proapoptotic Cardenolide of Cancer Cells That Up-Regulates Na,K-ATPase and Epithelial Tight Junctions

Sayonarah C. Rocha; Marco Túlio C. Pessôa; Luiza Dal-Rios Neves; Silmara L.G. Alves; Luciana Madeira da Silva; H. Santos; Soraya M.F. Oliveira; Alex Gutterres Taranto; Moacyr Comar; Isabella V. Gomes; Fábio Vieira dos Santos; Natasha Paixão; Luis Eduardo M. Quintas; François Noël; Antônio F. Pereira; Ana Claudia Tessis; Natália Lins da Silva Gomes; Otacilio C. Moreira; Ruth Rincon-Heredia; Fernando de Pilla Varotti; Gustavo Blanco; José A. F. P. Villar; Rubén G. Contreras; Leandro Augusto Barbosa

Cardiotonic steroids are used to treat heart failure and arrhythmia and have promising anticancer effects. The prototypic cardiotonic steroid ouabain may also be a hormone that modulates epithelial cell adhesion. Cardiotonic steroids consist of a steroid nucleus and a lactone ring, and their biological effects depend on the binding to their receptor, Na,K-ATPase, through which, they inhibit Na+ and K+ ion transport and activate of several intracellular signaling pathways. In this study, we added a styrene group to the lactone ring of the cardiotonic steroid digoxin, to obtain 21-benzylidene digoxin (21-BD), and investigated the effects of this synthetic cardiotonic steroid in different cell models. Molecular modeling indicates that 21-BD binds to its target Na,K-ATPase with low affinity, adopting a different pharmacophoric conformation when bound to its receptor than digoxin. Accordingly, 21-DB, at relatively high µM amounts inhibits the activity of Na,K-ATPase α1, but not α2 and α3 isoforms. In addition, 21-BD targets other proteins outside the Na,K-ATPase, inhibiting the multidrug exporter Pdr5p. When used on whole cells at low µM concentrations, 21-BD produces several effects, including: 1) up-regulation of Na,K-ATPase expression and activity in HeLa and RKO cancer cells, which is not found for digoxin, 2) cell specific changes in cell viability, reducing it in HeLa and RKO cancer cells, but increasing it in normal epithelial MDCK cells, which is different from the response to digoxin, and 3) changes in cell-cell interaction, altering the molecular composition of tight junctions and elevating transepithelial electrical resistance of MDCK monolayers, an effect previously found for ouabain. These results indicate that modification of the lactone ring of digoxin provides new properties to the compound, and shows that the structural change introduced could be used for the design of cardiotonic steroid with novel functions.


Cancer Research | 2017

Interplay between Immune Checkpoint Proteins and Cellular Metabolism

Sangbin Lim; Joshua B. Phillips; Luciana Madeira da Silva; Ming Zhou; Øystein Fodstad; Laurie B. Owen; Ming Tan

With the recent successes in immuno-oncology, renewed interest in the role of immune checkpoint modulators, such as the B7 family proteins, has escalated. The immune checkpoint proteins play a crucial role in the regulation of cellular immunity; however, their contribution to other aspects of cancer biology remains unclear. Accumulating evidence indicate that immune checkpoint proteins can regulate metabolic energetics of the tumor, the tumor microenvironment, and the tumor-specific immune response, leading to metabolic reprogramming of both malignant cells and immune cells involved in mounting and sustaining this response. Immune cell metabolism impacts the activation status of immune cells and ultimately the immune response in cancer. Tumor cells may deplete nutrients that immune cells require for optimal generation, expansion, and function. They may also generate toxic metabolites in the microenvironment or induce conserved inhibitory pathways that impair immune function and thus inhibit antitumor responses. In this review, we will discuss how cancer cells with altered expression of immune checkpoint proteins can potently inhibit immune function through the alteration of cellular and microenvironmental metabolism, providing a new perspective on the interplay between these pathways and offering a potential therapeutic intervention strategy in the treatment of malignant disease. Cancer Res; 77(6); 1245-9. ©2017 AACR.


Gynecologic Oncology | 2016

Unraveling the etiology of ovarian cancer racial disparity in the Deep South: Is it nature or nurture?

Jerlinda Ross; Katelyn V. Braswell; Luciana Madeira da Silva; Frances Mujica; Sam Stutsman; Michael A. Finan; William Nicolson; Mary Danner Harmon; Megan Missanelli; Alex Cohen; Ajay P. Singh; J.M. Scalici; Rodney P. Rocconi

BACKGROUND Our objective was to evaluate racial treatment and survival disparities in black women with ovarian cancer in the Deep South and to determine how environmental factors / socioeconomic status (SES) influence survival. METHODS A retrospective study of ovarian cancer patients from 2007 to 2014 was performed. Socioeconomic status (SES) was obtained though U.S. Census block data and compared using Yost scores. Comparisons were performed using standard statistical approaches. RESULTS A total of 393 patients were evaluated, 325 (83%) white and 68 (17%) black. Demographic information and surgical approach were similar in each racial group. However, compared to whites, black patients had lower rates of optimal debulking [89% vs. 71%, respectively (p=0.001)] and intraperitoneal chemotherapy (19% vs. 11%, p=0.01). Black women had lower SES parameters including education, income, and poverty. As a result, more black patients had the lowest SES (SES-1) when compared to white patients (17% vs. 41%, p<0.001). When controlling for these factors by cox regression analysis, a survival disadvantage was seen in black women for both progression free survival (16 vs. 27months, p=0.003) and overall survival (42 vs. 88months, p<0.001). CONCLUSIONS Despite controlling for clinical and environmental factors, a survival disadvantage was still observed in black patients with ovarian cancer in the Deep South. Black women had lower optimal debulking rates and more platinum resistant disease. These data suggest other factors like tumor biology may play a role in racial survival differences, however, more research is needed to determine this causation.


Medicinal Chemistry Research | 2017

Synthesis and in vitro evaluation of novel triazole/azide chalcones

Fernanda Cristina Gontijo Evangelista; Maralice O. Bandeira; Graziele Diniz da Silva; Marina Goulart da Silva; Silmara Nunes Andrade; Deisielly Ribeiro Marques; Luciana Madeira da Silva; Whocely Victor de Castro; Fábio Vieira dos Santos; Gustavo Henrique Ribeiro Viana; José A. F. P. Villar; Adriano de Paula Sabino; Fernando de Pilla Varotti

A series of 30 novel triazole/azide chalcone derivatives were synthesized by Claisen-Schmidt and Cu(I)-catalyzed cycloaddition reactions. The antiproliferative activity of each compound was evaluated against HeLa, RKO-AS45-1 and Wi-26VA4 cell lines. Terminal deoxynucleotidyl transferase dUTP nick end labeling assays indicated that compounds 4j and 5j significantly reduced the HeLa and RKO-AS45-1cell populations compared to the controls. The relative expression of the TP53 gene revealed changes in both cell lines after exposure to compounds 5j and 4j. The increased expression of the TP53 gene suggests a cellular attempt to repair DNA damage and indicates these triazole/azide chalcone derivatives as promising anticancer agents.


Cytotechnology | 2017

Digoxin reduces the mutagenic effects of Mitomycin C in human and rodent cell lines

Júlia Teixeira de Oliveira; Maria Cristina da Silva Barbosa; Luiz Fernando de Camargos; Isabella Viana Gomes da Silva; Fernando de Pilla Varotti; Luciana Madeira da Silva; Leonardo Marmo Moreira; Juliana P. Lyon; Vanessa J. S. V. Santos; Fábio Vieira dos Santos

Abstract Digoxin is a drug widely used to treat heart failure and studies have demonstrated its potential as anticancer agent. In addition, digoxin presents the potential to interact with a series of other compounds used in medicine. The aim of the present study was to evaluate in vitro the cytotoxicity, genotoxicity and mutagenicity of digoxin and its potential to interact with the mutagen Mitomycin C (MMC). The cytotoxicity of digoxin was assessed by employing the MTT method and the comet assay was performed to assess the genotoxicity of this medicine in CHO-K1 and HeLa cell lines. Besides, the cytokinesis-block micronucleus assay was performed to assess the mutagenicity and the antimutagenicity of this drug. The Ames assay was also performed with TA98 and TA100 strains of S. typhimurium. Results showed that digoxin was cytotoxic, genotoxic and mutagenic for HeLa and CHO-K1 cell lines at concentrations many times higher than those observed in human therapeutic conditions. Nevertheless, an antimutagenic effect against the mutagen MMC was observed on both cell lines in concentrations near those used therapeutically in humans. This chemoprotective effect observed is an interesting finding that should be better explored regarding its impact in anticancer chemotherapy.


Bioorganic & Medicinal Chemistry | 2015

γ-Benzylidene digoxin derivatives synthesis and molecular modeling: Evaluation of anticancer and the Na,K-ATPase activity effect.

Silmara L.G. Alves; Natasha Paixão; Letícia Gonçalves Resende Ferreira; Felipe R. S. Santos; Luiza Dal-Rios Neves; Gisele Capanema de Oliveira; Vanessa Faria Cortes; Kahlil Schwanka Salomé; Andersson Barison; Fábio Vieira dos Santos; Gisele Cenzi; Fernando de Pilla Varotti; Soraya M.F. Oliveira; Alex Gutterres Taranto; Moacyr Comar; Luciana Madeira da Silva; François Noël; Luis Eduardo M. Quintas; Leandro Augusto Barbosa; José A. F. P. Villar

Cardiotonic steroids (CS), natural compounds with traditional use in cardiology, have been recently suggested to exert potent anticancer effects. However, the repertoire of molecules with Na,K-ATPase activity and anticancer properties is limited. This paper describes the synthesis of 6 new digoxin derivatives substituted (on the C17-butenolide) with γ-benzylidene group and their cytotoxic effect on human fibroblast (WI-26 VA4) and cancer (HeLa and RKO) cell lines as well as their effect on Na,K-ATPase activity and expression. As digoxin, compound BD-4 was almost 100-fold more potent than the other derivatives for cytotoxicity with the three types of cells used and was also the only one able to fully inhibit the Na,K-ATPase of HeLa cells after 24h treatment. No change in the Na,K-ATPase α1 isoform protein expression was detected. On the other hand it was 30-40 fold less potent for direct Na,K-ATPase inhibition, when compared to the most potent derivatives, BD-1 and BD-3, and digoxin. The data presented here demonstrated that the anticancer effect of digoxin derivatives substituted with γ-benzylidene were not related with their inhibition of Na,K-ATPase activity or alteration of its expression, suggesting that this classical molecular mechanism of CS is not involved in the cytotoxic effect of our derivatives.


Cancer Research | 2016

Abstract 331: Beta-catenin nuclear translocation in colorectal cancer cells is suppressed by PDE10A inhibition, cGMP elevation, and activation of PKG

Kevin Lee; Ashley S. Lindsey; Luciana Madeira da Silva; Alisa Trinh; Bernard D. Gary; Joel Andrews; Veronica Ramirez-Alcantara; Adam B. Keeton; Wen-Chi L. Chang; Margie L. Clapper; Gary A. Piazza

Phosphodiesterase 10A (PDE10A) is a cGMP and cAMP degrading PDE isozyme that is highly expressed in the brain striatum where it appears to play an important role in cognition and psychomotor activity. PDE10 inhibitors are being developed for the treatment of schizophrenia and Huntington9s disease and are generally well tolerated, possibly because of low expression levels in most peripheral tissues. We recently reported high levels of PDE10 in colon tumors and that genetic silencing of PDE10 by siRNA or inhibition with small molecule inhibitors can suppress colon tumor cell growth with a high degree of selectivity over normal colonocytes (Li et al., Oncogene 2015). These observations suggest PDE10 may have an unrecognized role in tumorigenesis. Here we report that the concentration range by which the highly specific PDE10 inhibitor, Pf-2545920 (MP-10), inhibited colon tumor cell growth parallels the concentration range required to increase cGMP and cAMP levels, and activate PKG and PKA, respectively. Moreover, PDE10 knockdown by shRNA reduced the sensitivity of colon tumor cells to the growth inhibitory activity of Pf-2545920. Pf-2545920 also inhibited the translocation of β-catenin to the nucleus, thereby reducing β-catenin mediated transcription of survivin, which resulted in caspase activation and apoptosis. This was determined to be through a PKG mediated pathway through the use of small molecule inhibitors of PKG and PKA. PDE10 mRNA was also found to be elevated in colon tumors compared with normal tissues in a cDNA array. We also report the increase in PDE10 mRNA in 50% of a small collection of human clinical specimens collected at the Mitchell Cancer Institute (n = 13). In addition, novel PDE10 inhibitor, MCI-030, reduced tumor size and activated PDE10 signaling mechanisms in vivo. These findings suggest that PDE10 can be targeted for cancer therapy or prevention whereby inhibition results in cGMP elevation and PKG activation to reduce β-catenin-mediated transcription of survival proteins leading to the selective apoptosis of cancer cells. Citation Format: Kevin J. Lee, Ashley S. Lindsey, Luciana Madeira da Silva, Alisa Trinh, Bernard Gary, Joel Andrews, Veronica Ramirez-Alcantara, Adam B. Keeton, Wen-Chi Chang, Margie Clapper, Gary A. Piazza. Beta-catenin nuclear translocation in colorectal cancer cells is suppressed by PDE10A inhibition, cGMP elevation, and activation of PKG. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 331.

Collaboration


Dive into the Luciana Madeira da Silva's collaboration.

Top Co-Authors

Avatar

Gary A. Piazza

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Adam B. Keeton

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Kevin Lee

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Rodney P. Rocconi

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Bing Zhu

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xi Chen

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

Fernando de Pilla Varotti

Universidade Federal de São João del-Rei

View shared research outputs
Top Co-Authors

Avatar

Ashley S. Lindsey

University of South Alabama

View shared research outputs
Top Co-Authors

Avatar

J.M. Scalici

University of South Alabama

View shared research outputs
Researchain Logo
Decentralizing Knowledge