Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Inorg Biochem ; 168: 90-97, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28064102

RESUMO

Solid tumours are constituted of tumour cells, healthy cells recruited from the host tissues and soluble factors released by both these cell types. The present investigation examines the capacity of co-cultures between the HCEC colon epithelial cells and the HCT-116 colorectal cancer cells (mimicking the primary site of tumour growth) and between IHH hepatocytes and the HCT-116 colorectal cancer cells (metastatic site) to influence the effects of NAMI-A (imidazolium trans-imidazoledimethylsulphoxidetetrachloro ruthenate) on the tumour cells themselves. The growth of HCT-116 cells is significantly influenced when the cancer cells are sown on a monolayer of HCEC. The release of soluble factors by the healthy cells promotes, in HCT-116 colorectal cancer cells, the transcription of genes involved in growth, invasion and migration. NAMI-A is not cytotoxic to HCT-116 cells grown on plastics or co-cultured with HCEC or IHH cells, and maintains its ability to control the cell pseudo-metastatic ability, mimicked by the migration in the scratch test. The effects of NAMI-A on HCT-116 migration are supported by its inhibition of the transcription of the ABL-2, ATF-3 and RND-1 genes. In conclusion the study highlights the need of test systems more complex than a single cancer cell culture to study an anticancer drug in vitro and reinforces the hypothesis that NAMI-A targets the ability of the cancer cell to interact with the tumour microenvironment and with the signals that support its metastatic ability.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Dimetil Sulfóxido/análogos & derivados , Compostos Organometálicos/farmacologia , Microambiente Tumoral/fisiologia , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Dimetil Sulfóxido/química , Dimetil Sulfóxido/farmacologia , Células Epiteliais/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Hepatócitos/efeitos dos fármacos , Humanos , Compostos Organometálicos/química , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Compostos de Rutênio
2.
J Inorg Biochem ; 160: 225-35, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26961176

RESUMO

NAMI-A, imidazolium trans-imidazoledimethylsulfoxidetetrachlororuthenate, is a ruthenium-based drug characterised by the selective activity against tumour metastases. Previously we have shown the influence of the hepatic microenvironment to direct the arrest of the metastatic cells of colorectal cancer. Here we used the experimental model of HCT-116 colorectal cancer cells in vitro to explore whether the interference with α5ß1 integrin may mechanistically explain the anti-metastatic effect of NAMI-A. NAMI-A inhibits two important steps of the tumour metastatic progression of colorectal cancer, i.e. the adhesion and migration of the tumour cells on the extracellular matrix proteins. The fibronectin receptor α5ß1 integrin is likely involved in the anti-adhesive effects of NAMI-A on the HCT-116 colorectal cancer cells during their interaction with the extracellular matrix. Mechanistically, NAMI-A decreases the α5ß1 integrin expression, and reduces FAK (Focal Adhesion Kinase) auto-phosphorylation on Tyr397, an important signalling event, involved in α5ß1 integrin activation. These effects were validated by siRNA-induced knock down of the α5 integrin subunit and/or by the use of specific blocking mAbs against the active site of the integrin. Our results demonstrate the relevance of α5ß1 integrin for colorectal cancer. We also show that the anti-metastatic effect of NAMI-A depends on the modulation of this integrin. Thus, our data on NAMI-A support the new concept that metal-based drugs can inhibit tumour metastases through targeting of integrins and of other proteins which mediate tumour progression-related cell functions such as adhesion and migration.


Assuntos
Antineoplásicos/farmacologia , Dimetil Sulfóxido/análogos & derivados , Quinase 1 de Adesão Focal/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Integrina alfa5beta1/antagonistas & inibidores , Compostos Organometálicos/farmacologia , Rutênio/química , Anticorpos Neutralizantes/farmacologia , Antineoplásicos/química , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Dimetil Sulfóxido/química , Dimetil Sulfóxido/farmacologia , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Células HCT116 , Humanos , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Compostos Organometálicos/química , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Compostos de Rutênio
3.
Metallomics ; 7(10): 1439-50, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26338673

RESUMO

Gene expression profiling has been introduced into drug development to understand the activity of chemical entities in pre-clinical settings. The present study investigates the changes induced in gene expression by the ruthenium-based compound NAMI-A. The genes differentially expressed by NAMI-A are evaluated through whole-transcriptome analysis and RNA-sequencing in the metastatic MDA-MB-231 mammary carcinoma cells, in comparison to the non-tumorigenic HBL-100 mammary gland cells. NAMI-A treatment rapidly induces a relevant gene up-regulation that quickly returns to normal values. These changes differ between MDA-MB-231 and HBL-100 cells, highlighting the selectivity of the NAMI-A induced transcriptional perturbation in the invasive rather than in the non-tumorigenic cells. The transcriptional response, in the invasive MDA-MB-231 cells, comprises a set of early-response transcription factors and reveals a pharmacological signature in good agreement with the most peculiar NAMI-A behavior as a metastasis inhibitor such as cell cycle regulation and ECM remodeling. Globally, the results of this study indicate some transcription factors influencing the expression and activity of many downstream genes and proteins fundamentally involved in the functional effects of NAMI-A in vitro and in vivo.


Assuntos
Neoplasias da Mama/genética , Dimetil Sulfóxido/análogos & derivados , Compostos Organometálicos/farmacologia , Transcriptoma/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Dimetil Sulfóxido/farmacologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Compostos de Rutênio
4.
J Cell Biochem ; 116(10): 2385-96, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25857463

RESUMO

Colorectal cancer (CRC) metastasis dissemination to secondary sites represents the critical point for the patient's survival. The microenvironment is crucial to cancer progression, influencing tumour cell behaviour by modulating the expression and activation of molecules such as integrins, the cell-extracellular matrix interacting proteins participating in different steps of the tumour metastatic process. In this work, we investigated the role of α5ß1 integrin and how the microenvironment influences this adhesion molecule, in a model of colon cancer progression to the liver. The culture medium conditioned by the IHH hepatic cell line, and the extracellular matrix (ECM) proteins, modulate the activation of α5ß1 integrin in the colon cancer cell line HCT-116, and drives FAK phosphorylation during the process of cell adhesion to fibronectin, one of the main components of liver ECM. In these conditions, α5ß1 modulates the expression/activity of another integrin, α2ß1, involved in the cell adhesion to collagen I. These results suggest that α5ß1 integrin holds a leading role in HCT-116 colorectal cancer cells adhesion to the ECM through the modulation of the intracellular focal adhesion kinase FAK and the α2ß1 integrin activity. The driving role of the tumour microenvironment on CRC dissemination, here detected, and described, strengthens and adds new value to the concept that α5ß1 integrin can be an appropriate and relevant therapeutic target for the control of CRC metastases.


Assuntos
Proliferação de Células/genética , Neoplasias Colorretais/genética , Receptores de Vitronectina/biossíntese , Microambiente Tumoral/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/patologia , Matriz Extracelular/genética , Matriz Extracelular/patologia , Fibronectinas/metabolismo , Quinase 1 de Adesão Focal/biossíntese , Quinase 1 de Adesão Focal/genética , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Metástase Neoplásica , Fosforilação , Receptores de Vitronectina/genética
5.
Eur J Med Chem ; 95: 210-9, 2015 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-25817771

RESUMO

The proline-rich antibacterial peptide Bac7(1-35) protects mice against Salmonella typhimurium infection, despite its rapid clearance. To overcome this problem the peptide was linked to a polyethylene glycol (PEG) molecule either via a cleavable ester bond or via a non-hydrolysable amide bond. Both the PEGylated conjugates retained most of the in vitro activity against S. typhimurium. In addition, the ester bond was cleaved in human serum or plasma, releasing a carboxymethyl derivative of Bac7(1-35) which accounts for a higher activity of this peptide with relative to the other, non-hydrolysable form. Both PEGylated peptides maintained the capacity of the unconjugated form to kill bacteria without permeabilizing the bacterial membranes, by penetrating into cells. They exploited the same transporter as unmodified Bac7(1-35), suggesting it has the capacity to internalize quite sizeable cargo if this is linked to Bac7 fragment. PEGylation allows the peptide to have a wide distribution in mice, and a slow renal clearance, indicating that this strategy would improve the bioavailability of Bac7, and in principle of other antimicrobial peptides. This can be an equally important issue to reducing cytotoxicity for therapeutic use of these antibacterials.


Assuntos
Peptídeos Catiônicos Antimicrobianos/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Rim/metabolismo , Polietilenoglicóis/química , Salmonelose Animal/tratamento farmacológico , Salmonella typhimurium/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/química , Peptídeos Catiônicos Antimicrobianos/farmacocinética , Humanos , Rim/efeitos dos fármacos , Taxa de Depuração Metabólica , Camundongos , Salmonelose Animal/microbiologia , Distribuição Tecidual
6.
Protein Pept Lett ; 21(4): 382-90, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24164261

RESUMO

Bac7(1-35) is an active fragment of the bovine cathelicidin antimicrobial peptide Bac7, which selectively inactivates Gram-negative bacteria both in vitro and in mice infected with Salmonella typhimurium. It has a non-lytic mechanism of action, is rapidly internalized by susceptible bacteria and mammalian cells and likely acts by binding to internal targets. In this study we show that Bac7(1-35) accumulates selectively within primed macrophages with respect to resting monocytes. Confocal microscopy analysis showed that the peptide mainly distributes in the cytoplasm and perinuclear region of macrophages within 3 hours of incubation, without affecting cell viability. Cytotoxicity studies showed that the peptide does not induce necrotic or apoptotic damage up to concentrations 50-100-fold higher than minimal inhibitory concentrations (MIC). Moreover, Bac7(1-35) did not affect the ability of macrophages to engulf S. typhimurium, a species that may proliferate within this cell type. Conversely, when added to macrophages after phagocytosis, Bac7(1-35) caused a significant reduction in the number of recovered bacteria, indicating that it can kill the engulfed microorganisms directly and/or indirectly, via activation of the defense response of the cells.


Assuntos
Anti-Infecciosos/farmacologia , Anti-Infecciosos/farmacocinética , Macrófagos/microbiologia , Peptídeos Cíclicos/farmacocinética , Infecções por Salmonella/tratamento farmacológico , Salmonella typhimurium/efeitos dos fármacos , Animais , Bovinos , Linhagem Celular , Células Cultivadas , Humanos , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Peptídeos Cíclicos/farmacologia , Fagocitose
7.
BMC Microbiol ; 10: 178, 2010 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-20573188

RESUMO

BACKGROUND: Bac7 is a proline-rich peptide with a potent in vitro antimicrobial activity against Gram-negative bacteria. Here we investigated its activity in biological fluids and in vivo using a mouse model of S. typhimurium infection. RESULTS: The efficacy of the active 1-35 fragment of Bac7 was assayed in serum and plasma, and its stability in biological fluids analyzed by Western blot and mass spectrometry. The ability of the peptide to protect mice against Salmonella was assayed in a typhoid fever model of infection by determination of survival rates and bacterial load in liver and spleen of infected animals. In addition, the peptide's biodistribution was evaluated by using time-domain optical imaging. Bac7(1-35) retained a substantial in vivo activity showing a very low toxicity. The peptide increased significantly the number of survivors and the mean survival times of treated mice reducing the bacterial load in their organs despite its rapid clearance. CONCLUSIONS: Our results provide a first indication for a potential development of Bac7-based drugs in the treatment of salmonellosis and, eventually, other Gram-negative infections. The in vivo activity for this peptide might be substantially enhanced by decreasing its excretion rate or modifying the treatment schedule.


Assuntos
Antibacterianos/farmacologia , Peptídeos Catiônicos Antimicrobianos/farmacologia , Salmonelose Animal/tratamento farmacológico , Salmonella typhimurium , Animais , Peptídeos Catiônicos Antimicrobianos/farmacocinética , Fígado/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Salmonelose Animal/microbiologia , Baço/microbiologia , Distribuição Tecidual
8.
Biomacromolecules ; 10(6): 1429-35, 2009 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-19405545

RESUMO

In this work we study (i) the formation and stabilization of silver nanoparticles in a bioactive chitosan-derived polysaccharide solution, (ii) the antimicrobial properties, either in solution or in 3D hydrogel structures, obtained by mixtures with the polysaccharide alginate, and (iii) the cytotoxicity of the latter nanocomposite materials on different eukaryotic cell lines. Antimicrobial results show that these nanocomposite systems display a very effective bactericidal activity toward both Gram+ and Gram- bacteria. However, the hydrogel does not show any cytotoxic effect toward three different eukaryotic cell lines. This is due to the fact that the nanoparticles, immobilized in the gel matrix, can exert their antimicrobial activity by simple contact with the bacterial membrane, while they can not be uptaken and internalized by eukaryotic cells. This novel finding could advantageously contribute to responding to the growing concerns on the toxicity of nanoparticles and facilitate the use of silver-biopolymer composites in the preparation of biomaterials.


Assuntos
Antibacterianos/farmacologia , Nanopartículas Metálicas , Prata/química , Antibacterianos/química , Escherichia coli/efeitos dos fármacos , Citometria de Fluxo , Hidrogéis , Espectrometria de Massas , Testes de Sensibilidade Microbiana , Microscopia Eletrônica de Transmissão , Pseudomonas aeruginosa/efeitos dos fármacos , Espectrofotometria Ultravioleta , Staphylococcus aureus/efeitos dos fármacos
9.
Biomacromolecules ; 10(2): 210-3, 2009 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-19140690

RESUMO

Alditol bearing chitosans have shown the ability to reduce silver ions in mild conditions and without addition of exogenous reducing agents. The ion reduction induces the formation of a lactone moiety on the polysaccharide (Fetizon reaction) without causing C-C bond cleavage on the polyol. The close and multivalent arrangement of the endogenous reducing agent (alditols) on the polysaccharide backbone resulted in the formation of silver nanoparticles (phi < 10 nm), which induced a considerable SERS effect and led to hydrogel formation.


Assuntos
Nanopartículas Metálicas/química , Prata , Polímeros/química , Polissacarídeos/química , Substâncias Redutoras , Álcoois Açúcares/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...