EMERSON SOARES BERNARDES

Resumo

Bachelor's at Farmácia from Universidade Federal de Ouro Preto (1998) and doctorate at Applied Imunology from Universidade de São Paulo (2004). Has experience in Medicine, acting on the following subjects: galectina-3, carboidratos, trypanosoma cruzi, carcinogênesis and macrophage. (Text obtained from the Currículo Lattes on October 8th 2021)


Possui graduação em Farmácia pela Universidade Federal de Ouro Preto (1998), mestrado e doutorado em Imunologia Básica e Aplicada pela Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (2004), com período de Doutorado Sanduíche pela Universidade da California, Davis, USA. Realizou pós-doutoramento durante o período de 2004 a 2008 pela Faculdade de Medicina da USP-Ribeirão Preto. Trabalhou como pesquisador contratado pelo Instituto de Patologia e Imunologia Molecular da Universidade do Porto - IPATIMUP em Portugal no período de 2008 a 2011. Retornou ao Brasil como pesquisador visitante na Faculdade de Medicina da USP - São Paulo (2011-2012) e foi posteriormente contratado como pesquisador no Instituto do Câncer do Estado de São Paulo (2012-2013). Coordenou um projeto Jovem Pesquisador financiado pela FAPESP (2012-2016 - Desenvolvimento e Produção de Radiofármacos Emissores de Pósitrons com Aplicações Diagnósticas em Oncologia) e está integrado como pesquisador Colaborador no Instituto de Pesquisas Energéticas e Nucleares (IPEN). Tem atuado na área da Glicobiologia, com ênfase na participação de proteínas ligantes de carboidratos em processos inflamatórios e no Câncer. Atualmente é professor do Programa de Pós-Graduação do IPEN-USP Tecnologia Nuclear - Aplicações, tem experiência na área de Radiofarmácia, com ênfase no desenvolvimento de Radiofármacos inéditos para diagnóstico e terapia em Oncologia. (Texto extraído do Currículo Lattes em 08 out. 2021)

Projetos de Pesquisa
Unidades Organizacionais
Cargo

Resultados de Busca

Agora exibindo 1 - 9 de 9
  • Artigo IPEN-doc 29303
    Superior Machine Learning Method for breast cancer cell lines identification
    2022 - FAROOQ, SAJID; CARAMEL-JUVINO, AMANDA; DEL-VALLE, MATHEUS; SANTOS, SOFIA; BERNARDES, EMERSON S.; ZEZELL, DENISE M.
    We propose an artificial intelligence platform based on machine learning (ML) algorithm using Neighborhood Component analysis and K-Nearest Neighbors for breast cancer cell lines recognition. Our model presents up to 97% accuracy for identification of breast cancer cell lines.
  • Artigo IPEN-doc 28362
    Synthesis, biological evaluation and molecular docking studies of novel 1,2,3-triazole-quinazolines as antiproliferative agents displaying ERK inhibitory activity
    2021 - NUNES, PAULO S.G.; SILVA, GABRIEL da; NASCIMENTO, SOFIA; MANTOANI, SUSIMAIRE P.; ANDRADE, PETERSON de; BERNARDES, EMERSON S.; KAWANO, DANIEL F.; LEOPOLDINO, ANDREIA M.; CARVALHO, IVONE
    ERK1/2 inhibitors have attracted special attention concerning the ability of circumventing cases of innate or log-term acquired resistance to RAF and MEK kinase inhibitors. Based on the 4-aminoquinazoline pharmacophore of kinases, herein we describe the synthesis of 4-aminoquinazoline derivatives bearing a 1,2,3-triazole stable core to bridge different aromatic and heterocyclic rings using copper-catalysed azide-alkyne cycloaddition reaction (CuAAC) as a Click Chemistry strategy. The initial screening of twelve derivatives in tumoral cells (CAL-27, HN13, HGC-27, and BT-20) revealed that the most active in BT-20 cells (25a, IC50 24.6 μM and a SI of 3.25) contains a more polar side chain (sulfone). Furthermore, compound 25a promoted a significant release of lactate dehydrogenase (LDH), suggesting the induction of cell death by necrosis. In addition, this compound induced G0/G1 stalling in BT-20 cells, which was accompanied by a decrease in the S phase. Western blot analysis of the levels of p-STAT3, p-ERK, PARP, p53 and cleaved caspase-3 revealed p-ERK1/2 and p-STA3 were drastically decreased in BT-20 cells under 25a incubation, suggesting the involvement of these two kinases in the mechanisms underlying 25a-induced cell cycle arrest, besides loss of proliferation and viability of the breast cancer cell. Molecular docking simulations using the ERK-ulixertinib crystallographic complex showed compound 25a could potentially compete with ATP for binding to ERK in a slightly higher affinity than the reference ERK1/2 inhibitor. Further in silico analyses showed comparable toxicity and pharmacokinetic profiles for compound 25a in relation to ulixertinib.
  • Resumo IPEN-doc 27521
  • Artigo IPEN-doc 26869
    Engineering of galectin-3 for glycan-binding optical imaging
    2020 - DE LEO, THAIS C.; SANTOS, SOFIA N. dos; ANDRADE, CAMILLO D.C.; RICCI, EDUARDO; TURATO, WALTER M.; LOPES, NORBERTO P.; OLIVEIRA, RALPH S.; BERNARDES, EMERSON S.; DIAS-BARUFFI, MARCELO
    Galectin-3 (Gal-3) is a multifunctional glycan-binding protein that participates in many pathophysiological events and has been described as a biomarker and potential therapeutic target for severe disorders, such as cancer. Several probes for Gal-3 or its ligands have been developed, however both the pathophysiological mechanisms and potential biomedical applications of Gal-3 remain not fully assessed. Molecular imaging using bioluminescent probes provides great sensitivity for in vivo and in vitro analysis for both cellular and whole multicellular organism tracking and target detection. Here, we engineered a chimeric molecule consisting of Renilla luciferase fused with mouse Gal-3 (RLuc-mGal-3). RLuc-mGal-3 preparation was highly homogenous, soluble, active, and has molecular mass of 65,870.95 Da. This molecule was able to bind to MKN45 cell surface, property which was inhibited by the reduction of Gal-3 ligands on the cell surface by the overexpression of ST6GalNAc-I. In order to obtain an efficient and stable delivery system, RLuc-mGal-3 was adsorbed to poly-lactic acid nanoparticles, which increased binding to MKN45 cells in vitro. Furthermore, bioluminescence imaging showed that RLuc-mGal-3 was able to indicate the presence of implanted tumor in mice, event drastically inhibited by the presence of lactose. This novel bioluminescent chimeric molecule offers a safe and highly sensitive alternative to fluorescent and radiolabeled probes with potential application in biomedical research for a better understanding of the distribution and fate of Gal-3 and its ligands in vitro and in vivo.
  • Artigo IPEN-doc 26663
    Galectin-3 regulates the expression of tumor glycosaminoglycans and increases the metastatic potential of breast cancer
    2019 - PEREIRA, JONATHAS X.; SANTOS, SOFIA N. dos; PEREIRA, THAIS C.; CABANEL, MARIANA; CHAMMAS, ROGER; OLIVEIRA, FELIPE L. de; BERNARDES, EMERSON S.; EL-CHEIKH, MARCIA C.
    Galectin-3 (Gal-3) is a multifunctional β-galactoside-binding lectin that once synthesized is expressed in the nucleus, cytoplasm, cell surface, and extracellular environment. Gal-3 plays an important role in breast cancer tumors due to its ability to promote interactions between cell-cell and cell-extracellular matrix (ECM) elements, increasing tumor survival and metastatic dissemination. Still, the mechanism by which Gal-3 interferes with tumor cell migration and metastasis formation is complex and not fully understood. Here, we showed that Gal-3 knockdown increased the migration ability of 4T1 murine breast cancer cells in vitro. Using the 4T1 orthotopic breast cancer spontaneous metastasis mouse model, we demonstrated that 4T1-derived tumors were significantly larger in the presence of Gal-3 (scramble) in comparison with Gal-3 knockdown 4T1-derived tumors. Nevertheless, Gal-3 knockdown 4T1 cells were outnumbered in the bone marrow in comparison with scramble 4T1 cells. Finally, we reported here a decrease in the content of cell-surface syndecan-1 and an increase in the levels of chondroitin sulfate proteoglycans such as versican in Gal-3 knockdown 4T1 cells both in vitro and in vivo. Overall, our findings establish that Gal-3 downregulation during breast cancer progression regulates cell-associated and tumor microenvironment glycosaminoglycans (GAGs)/proteoglycans (PG), thus enhancing the metastatic potential of tumor cells.
  • Artigo IPEN-doc 26479
    Octreotide nanoparticles showed affinity for in vivo MIA Paca-2 inducted pancreas ductal adenocarcinoma mimicking pancreatic polypeptide-secreting tumor of the distal pancreas (PPoma)
    2019 - BRAGA, THAIS L.; PINTO, SUYENE R.; REIS, SARA R.R. dos; PORTILHO, FILIPE L.; BARROS, ALINE O. da S. de; BERNARDES, EMERSON S.; SANTOS, SOFIA N. dos; ALENCAR, LUCIANA M.R.; RICCI-JUNIOR, EDUARDO; SANTOS-OLIVEIRA, RALPH
    Purpose Pancreatic Polypeptide-secreting tumor of the distal pancreas (PPoma) is a rare, difficult and indolent type of cancer with a survival rate of 5-year in only 10% of all cases. The PPoma is classified as a neuroendocrine tumor (NET) not functioning that overexpresses SSTR 2 (somatostatin receptor subtype 2). Thus, in order to improve the diagnosis of this type of tumor, we developed nanoparticulate drug carriers based on poly-lactic acid (PLA) polymer loaded with octreotide and radiolabeled with Technetium-99 m (99mTc). Methods PLA/PVA octreotide nanoparticles were developed by double-emulsion technique. These nanoparticles were characterized by Atomic Force Microscopy (AFM) and Dynamic Light Scattering (DLS) and radiolabeled with 99mTc by the direct via forming 99mTc-PLA/PVA octreotide nanoparticles. The safety of these nanosystems was evaluated by the MTT cell toxicity assay and their in vivo biodistribution was evaluated in xenografted inducted animals. Results The results showed that a 189 nm sized nanoparticle were formed with a PDI of 0,097, corroborating the monodispersive behavior. These nanoparticles were successfully radiolabeled with 99mTc showing uptake by the inducted tumor. The MTT assay corroborated the safety of the nanosystem for the cells. Conclusion The results support the use of this nanosystem (99mTc-PLA/PVA octreotide nanoparticles) as imaging agent for PPoma.
  • Artigo IPEN-doc 24361
    MUC1 aptamer-capped mesoporous silica nanoparticles for controlled drug delivery and radio-imaging applications
    2017 - PASCUAL, LLUIS; CERQUEIRA-COUTINHO, CRISTAL; GARCIA-FERNANDEZ, ALBA; LUIS, BEATRIZ de; BERNARDES, EMERSON S.; ALBERNAZ, MARTA S.; MISSAILIDIS, SOTIRIS; MARTINEZ-MANEZ, RAMON; SANTOS-OLIVEIRA, RALPH; ORZAEZ, MAR; SANCENON, FELIX
    Mucin 1 (MUC1) is a cell surface protein overexpressed in breast cancer. Mesoporous silica nanoparticles (MSNs) loaded with safranin O, functionalized with aminopropyl groups and gated with the negatively charged MUC1 aptamer have been prepared (S1-apMUC1) for specific targeting and cargo release in tumoral versus non-tumoral cells. Confocal microscopy studies showed that the S1-apMUC1 nanoparticles were internalized in MDA-MB-231 breast cancer cells that overexpress MUC1 receptor with subsequent pore opening and cargo release. Interestingly, the MCF-10-A non-tumorigenic breast epithelial cell line that do not overexpress MUC1, showed reduced (S1- apMUC1) internalization. Negligible internalization was also found for S1-ap nanoparticles that contained a scrambled DNA sequence as gatekeeper. S2-apMUC1 nanoparticles (similar to S1-apMUC1 but loaded with doxorubicin) internalized in MDA-MB-231 cells and induced a remarkable reduction in cell viability. Moreover, S1-apMUC1 nanoparticles radio-labeled with 99mTc (S1-apMUC1-Tc) showed a remarkable tumor targeting in in vivo studies with MDA-MB-231 tumor-bearing Balb/c mice. © 2017 Elsevier Inc. All rights reserved.
  • Artigo IPEN-doc 23199
    Galectin-3 acts as an angiogenic switch to induce tumor angiogenesis via Jagged-1/Notch activation
    2017 - SANTOS, SOFIA N. dos; SHELDON, HELEN; PEREIRA, JONATHAS X.; PALUCH, CHRISTOPHER; BRIDGES, ESTHER M.; EL-CHEIKH, MARCIA C.; HARRIS, ADRIAN L.; BERNARDES, EMERSON S.
    Angiogenesis is a coordinated process tightly regulated by the balance between Delta-like-4 (DLL4) and Jagged-1 (JAG1) in endothelial cells. Here we show that galectin-3 (gal-3), a glycan-binding protein secreted by cancer cells under hypoxic conditions, triggers sprouting angiogenesis, assisted by hypoxic changes in the glycosylation status of endothelial cells that enhance binding to gal-3. Galectin-3' s proangiogenic functions were found to be predominantly dependent on the Notch ligand JAG1. Differential direct binding to JAG1 was shown by surface plasmon resonance assay. Upon binding to Notch ligands, gal-3 preferentially increased JAG1 protein half-life over DLL4 and preferentially activated JAG1/Notch-1 signaling in endothelial cells. JAG1 overexpression in Lewis lung carcinoma cells accelerated tumor growth in vivo, but this effect was prevented in Lgals3(-/-) mice. Our findings establish gal-3 as a molecular regulator of the JAG1/Notch-1 signaling pathway and have direct implications for the development of strategies aimed at controlling tumor angiogenesis.
  • Artigo IPEN-doc 22660
    The deficiency of galectin-3 in stromal cells leads to enhanced tumor growth and bone marrow metastasis
    2016 - PEREIRA, JONATHAS X.; AZEREDO, MARIA C.B.; MARTINS, FELIPE S.; CHAMMAS, ROGER; OLIVEIRA, FELIPE L.; SANTOS, SOFIA N.; BERNARDES, EMERSON S.; EL-CHEIKH, MARCIA C.
    Background: Galectin-3 is a multifunctional β-galactoside-binding lectin that once synthesized, is expressed in the nucleus, cytoplasm, cell surface and in the extracellular environment. Because of its unique structure, galectin-3 can oligomerize forming lattice upon binding to multivalent oligossacharides and influence several pathologic events such as tumorigenesis, invasion and metastasis. Methods: In our study, balb/c Lgals3+/+ and Lgals3−/− female mice were inoculated in the fourth mammary fat pad with 4T1 breast cancer cell line. The primary tumor, inguinal lymph nodes and iliac bone marrow were evaluated 15, 21 and 28 days post-injection. The primary tumor growth was evaluated by measuring the external diameter, internal growth by ultrasound and weight of the excised tumor. The presence of cancer cells in the draining lymph nodes and iliac crest bone marrow were performed by immunohistochemistry, PCR and clonogenic metastatic assay. Results: In this study we demonstrated that the deletion of galectin-3 in the host affected drastically the in vivo growth rate of 4T1 tumors. The primary tumors in Lgals3−/− mice displayed a higher proliferative rate (p < 0,05), an increased necrotic area (p < 0,01) and new blood vessels with a wider lumen in comparison with tumors from Lgals3+/+ mice (P < 0,05). Moreover, we detected a higher number of 4T1-derived metastatic colonies in the lymph nodes and the bone marrow of Lgals3−/− mice (p < 0,05). Additionally, healthy Lgals3−/− control mice presented an altered spatial distribution of CXCL12 in the bone marrow, which may explain at least in part the initial colonization of this organ in Lgals3−/− injected with 4T1 cells. Conclusions: Taken together, our results demonstrate for the first time that the absence of galectin-3 in the host microenvironment favors the growth of the primary tumors, the metastatic spread to the inguinal lymph nodes and bone marrow colonization by metastatic 4T1 tumor cells.