Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Bioengineering (Basel) ; 9(11)2022 Nov 10.
Article in English | MEDLINE | ID: mdl-36354586

ABSTRACT

Extracellular vesicles (EVs) have been the focus of great attention over the last decade, considering their promising application as next-generation therapeutics. EVs have emerged as relevant mediators of intercellular communication, being associated with multiple physiological processes, but also in the pathogenesis of several diseases. Given their natural ability to shuttle messages between cells, EVs have been explored both as inherent therapeutics in regenerative medicine and as drug delivery vehicles targeting multiple diseases. However, bioengineering strategies are required to harness the full potential of EVs for therapeutic use. For that purpose, a good understanding of EV biology, from their biogenesis to the way they are able to shuttle messages and establish interactions with recipient cells, is needed. Here, we review the current state-of-the-art on EV biology, complemented by representative examples of EVs roles in several pathophysiological processes, as well as the intrinsic therapeutic properties of EVs and paradigmatic strategies to produce and develop engineered EVs as next-generation drug delivery systems.

2.
Biomolecules ; 12(7)2022 06 29.
Article in English | MEDLINE | ID: mdl-35883468

ABSTRACT

Despite its low abundance, phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is a key modulator of membrane-associated signaling events in eukaryotic cells. Temporal and spatial regulation of PI(4,5)P2 concentration can achieve localized increases in the levels of this lipid, which are crucial for the activation or recruitment of peripheral proteins to the plasma membrane. The recent observation of the dramatic impact of physiological divalent cation concentrations on PI(4,5)P2 clustering, suggests that protein anchoring to the plasma membrane through PI(4,5)P2 is likely not defined solely by a simple (monomeric PI(4,5)P2)/(protein bound PI(4,5)P2) equilibrium, but instead depends on complex protein interactions with PI(4,5)P2 clusters. The insertion of PI(4,5)P2-binding proteins within these clusters can putatively modulate protein-protein interactions in the membrane, but the relevance of such effects is largely unknown. In this work, we characterized the impact of Ca2+ on the organization and protein-protein interactions of PI(4,5)P2-binding proteins. We show that, in giant unilamellar vesicles presenting PI(4,5)P2, the membrane diffusion properties of pleckstrin homology (PH) domains tagged with a yellow fluorescent protein (YFP) are affected by the presence of Ca2+, suggesting direct interactions between the protein and PI(4,5)P2 clusters. Importantly, PH-YFP is found to dimerize in the membrane in the absence of Ca2+. This oligomerization is inhibited in the presence of physiological concentrations of the divalent cation. These results confirm that cation-dependent PI(4,5)P2 clustering promotes interactions between PI(4,5)P2-binding proteins and has the potential to dramatically influence the organization and downstream interactions of PI(4,5)P2-binding proteins in the plasma membrane.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate , Unilamellar Liposomes , Cations, Divalent/metabolism , Cell Membrane/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositols/metabolism , Unilamellar Liposomes/metabolism
3.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769158

ABSTRACT

Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is an essential plasma membrane component involved in several cellular functions, including membrane trafficking and cytoskeleton organization. This function multiplicity is partially achieved through a dynamic spatiotemporal organization of PI(4,5)P2 within the membrane. Here, we use a Förster resonance energy transfer (FRET) approach to quantitatively assess the extent of PI(4,5)P2 confinement within the plasma membrane. This methodology relies on the rigorous evaluation of the dependence of absolute FRET efficiencies between pleckstrin homology domains (PHPLCδ) fused with fluorescent proteins and their average fluorescence intensity at the membrane. PI(4,5)P2 is found to be significantly compartmentalized at the plasma membrane of HeLa cells, and these clusters are not cholesterol-dependent, suggesting that membrane rafts are not involved in the formation of these nanodomains. On the other hand, upon inhibition of actin polymerization, compartmentalization of PI(4,5)P2 is almost entirely eliminated, showing that the cytoskeleton network is the critical component responsible for the formation of nanoscale PI(4,5)P2 domains in HeLa cells.


Subject(s)
Cytoskeleton/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Cell Membrane/metabolism , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Membrane Microdomains/metabolism , Microscopy , Phosphatidylinositol 4,5-Diphosphate/analysis
4.
Biomedicines ; 9(9)2021 Sep 10.
Article in English | MEDLINE | ID: mdl-34572379

ABSTRACT

Peptides have been thoroughly studied as new therapeutic strategies for cancer treatment. In this work, we explored in vitro the anticancer potential of three novel peptides derived from the C-terminal of azurin, an anticancer bacterial protein produced by Pseudomonas aeruginosa. CT-p26, CT-p19 and CT-p19LC peptides were previously obtained through an in silico peptide design optimization process, CT-p19LC being the most promising as it presented higher hydrophobicity and solubility, positive total charge and, most importantly, greater propensity for anticancer activity. Therefore, in this study, through proliferation and apoptosis assays, CT-p19LC was tested in four cancer cell lines-A549, MCF-7, HeLa and HT-29-and in two non-cancer cell lines-16HBE14o- and MCF10A. Its membrane-targeting activity was further evaluated with zeta potential measurements and membrane order was assessed with the Laurdan probe. The results obtained demonstrated that CT-p19LC decreases cell viability through induction of cell death and binds to the plasma membrane of cancer cells, but not to non-cancer cells, making them less rigid. Overall, this study reveals that CT-p19LC is an auspicious selective anticancer peptide able to react with cancer cell membranes and cause effective action.

5.
J Control Release ; 337: 329-342, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34311024

ABSTRACT

Lung cancer is still the main cause of cancer-related deaths worldwide. Its treatment generally includes surgical resection, immunotherapy, radiotherapy, and chemo-targeted therapies such as the application of tyrosine kinase inhibitors. Gefitinib (GEF) is one of them, but its poor solubility in gastric fluids weakens its bioavailability and therapeutic activity. In addition, like all other chemotherapy treatments, GEF administration can cause damage to healthy tissues. Therefore, the development of novel GEF delivery systems to increase its bioavailability and distribution in tumor site is highly demanded. Herein, an innovative strategy for GEF delivery, by functionalizing PLGA nanoparticles with p28 (p28-NPs), a cell-penetrating peptide derived from the bacterial protein azurin, was developed. Our data indicated that p28 potentiates the selective interaction of these nanosystems with A549 lung cancer cells (active targeting). Further p28-NPs delivering GEF (p28-NPs-GEF) were able to selectively reduce the metabolic activity of A549 cells, while no impact was observed in non-tumor cells (16HBE14o-). In vivo studies using A549 subcutaneous xenograft showed that p28-NPs-GEF reduced A549 primary tumor burden and lung metastases formation. Overall, the design of a p28-functionalized delivery nanosystem to effectively penetrate the membranes of cancer cells while deliver GEF could provide a new strategy to improve lung cancer therapy.


Subject(s)
Antineoplastic Agents , Lung Neoplasms , Nanoparticles , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Gefitinib , Humans , Lung Neoplasms/drug therapy , Tumor Burden
6.
Cell Microbiol ; 23(8): e13340, 2021 08.
Article in English | MEDLINE | ID: mdl-33822465

ABSTRACT

Trimeric Autotransporter Adhesins (TAA) found in Gram-negative bacteria play a key role in virulence. This is the case of Burkholderia cepacia complex (Bcc), a group of related bacteria able to cause infections in patients with cystic fibrosis. These bacteria use TAAs, among other virulence factors, to bind to host protein receptors and their carbohydrate ligands. Blocking such contacts is an attractive approach to inhibit Bcc infections. In this study, using an antibody produced against the TAA BCAM2418 from the epidemic strain Burkholderia cenocepacia K56-2, we were able to uncover its roles as an adhesin and the type of host glycan structures that serve as recognition targets. The neutralisation of BCAM2418 was found to cause a reduction in the adhesion of the bacteria to bronchial cells and mucins. Moreover, in vivo studies have shown that the anti-BCAM2418 antibody exerted an inhibitory effect during infection in Galleria mellonella. Finally, inferred by glycan arrays, we were able to predict for the first time, host glycan epitopes for a TAA. We show that BCAM2418 favoured binding to 3'sialyl-3-fucosyllactose, histo-blood group A, α-(1,2)-linked Fuc-containing structures, Lewis structures and GM1 gangliosides. In addition, the glycan microarrays demonstrated similar specificities of Burkholderia species for their most intensely binding carbohydrates.


Subject(s)
Burkholderia Infections , Burkholderia cenocepacia , Adhesins, Bacterial , Bacterial Adhesion , Humans , Polysaccharides
7.
Sci Rep ; 11(1): 5624, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33707642

ABSTRACT

Burkholderia cenocepacia is known for its capacity of adherence and interaction with the host, causing severe opportunistic lung infections in cystic fibrosis patients. In this work we produced Giant Plasma Membrane Vesicles (GPMVs) from a bronchial epithelial cell line and validated their use as a cell-like alternative to investigate the steps involved in the adhesion process of B. cenocepacia. RNA-sequencing was performed and the analysis of the B. cenocepacia K56-2 transcriptome after the first contacts with the surface of host cells allowed the recognition of genes implicated in bacterial adaptation and virulence-associated functions. The sensing of host membranes led to a transcriptional shift that caused a cascade of metabolic and physiological adaptations to the host specific environment. Many of the differentially expressed genes encode proteins related with central metabolic pathways, transport systems, cellular processes, and virulence traits. The understanding of the changes in gene expression that occur in the early steps of infection can uncover new proteins implicated in B. cenocepacia-host cell adhesion, against which new blocking agents could be designed to control the progression of the infectious process.


Subject(s)
Bronchi/pathology , Burkholderia cenocepacia/genetics , Cell Membrane/microbiology , Epithelial Cells/microbiology , Transcriptome/genetics , Burkholderia cenocepacia/pathogenicity , Cell Adhesion , Cell Line , Cell Membrane/ultrastructure , Cluster Analysis , Epithelial Cells/ultrastructure , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Humans , Metabolic Networks and Pathways/genetics , Models, Biological
8.
Front Cell Dev Biol ; 8: 553444, 2020.
Article in English | MEDLINE | ID: mdl-33224943

ABSTRACT

Mesenchymal stromal cells (MSC) hold great promise for tissue engineering and cell-based therapies due to their multilineage differentiation potential and intrinsic immunomodulatory and trophic activities. Over the past years, increasing evidence has proposed extracellular vesicles (EVs) as mediators of many of the MSC-associated therapeutic features. EVs have emerged as mediators of intercellular communication, being associated with multiple physiological processes, but also in the pathogenesis of several diseases. EVs are derived from cell membranes, allowing high biocompatibility to target cells, while their small size makes them ideal candidates to cross biological barriers. Despite the promising potential of EVs for therapeutic applications, robust manufacturing processes that would increase the consistency and scalability of EV production are still lacking. In this work, EVs were produced by MSC isolated from different human tissue sources [bone marrow (BM), adipose tissue (AT), and umbilical cord matrix (UCM)]. A serum-/xeno-free microcarrier-based culture system was implemented in a Vertical-WheelTM bioreactor (VWBR), employing a human platelet lysate culture supplement (UltraGROTM-PURE), toward the scalable production of MSC-derived EVs (MSC-EVs). The morphology and structure of the manufactured EVs were assessed by atomic force microscopy, while EV protein markers were successfully identified in EVs by Western blot, and EV surface charge was maintained relatively constant (between -15.5 ± 1.6 mV and -19.4 ± 1.4 mV), as determined by zeta potential measurements. When compared to traditional culture systems under static conditions (T-flasks), the VWBR system allowed the production of EVs at higher concentration (i.e., EV concentration in the conditioned medium) (5.7-fold increase overall) and productivity (i.e., amount of EVs generated per cell) (3-fold increase overall). BM, AT and UCM MSC cultured in the VWBR system yielded an average of 2.8 ± 0.1 × 1011, 3.1 ± 1.3 × 1011, and 4.1 ± 1.7 × 1011 EV particles (n = 3), respectively, in a 60 mL final volume. This bioreactor system also allowed to obtain a more robust MSC-EV production, regarding their purity, compared to static culture. Overall, we demonstrate that this scalable culture system can robustly manufacture EVs from MSC derived from different tissue sources, toward the development of novel therapeutic products.

9.
Front Cell Dev Biol ; 8: 471, 2020.
Article in English | MEDLINE | ID: mdl-32733876

ABSTRACT

Recently, cell-based therapies have been explored as a strategy to enhance the specificity of anticancer therapeutic agents. In this perspective, human mesenchymal stromal cells (MSC) hold a promising future as cell delivery systems for anticancer proteins due to their unique biological features. In this study, we engineered human MSC to secrete a human codon-optimized version of azurin (hazu), a bacterial protein that has demonstrated anticancer activity toward different cancer models both in vitro and in vivo. To this end, microporation was used to deliver plasmid DNA encoding azurin into MSC derived from bone marrow (BM) and umbilical cord matrix (UCM), leading to expression and secretion of hazu to the conditioned medium (CM). Engineered hazu-MSC were shown to preserve tumor tropism toward breast (MCF-7) and lung (A549) cancer cell lines, comparable to non-modified MSC. Azurin was detected in the CM of transfected MSC and, upon treatment with hazu-MSC-CM, we observed a decrease in cancer cell proliferation, migration, and invasion, and an increase in cell death for both cancer cell lines. Moreover, expression of azurin caused no changes in MSC expression profile of cytokines relevant in the context of cancer progression, thus suggesting that the antitumoral effects induced by hazu-MSC secretome might be due to the presence of azurin independently. In conclusion, data shown herein indicate that MSC-produced azurin in a CM configuration elicits an anticancer effect.

10.
PLoS One ; 14(7): e0219900, 2019.
Article in English | MEDLINE | ID: mdl-31314780

ABSTRACT

Development of a healthy musculoskeletal system is of high concern for horse breeders and users. A longitudinal field study was performed in order to: (i) evaluate growth patterns and long-term changes on bone quality, bone metabolism, growth factors and metabolic variables in the Lusitano horse; and (ii) retrospectively assess whether these changes were related with radiographic findings regarding osteochondrosis-like lesions (OC) at the onset of training. Thirty-four Lusitano foals born and raised at four stud-farms, were periodically weighed (BW), and measured (withers height-WH) from birth to 36 months of age. On the same days, blood samples were collected for determination of osteocalcin, bone alkaline phosphatase, insulin-like growth factor I (IGF-I), leptin, insulin, glucose, parathyroid hormone (PTH), calcium, phosphorus and magnesium plasma concentrations, and quantitative ultrasound measurements were performed on the right third metacarpal bone (McIII). At the end of the study horses underwent radiographic examination of the four fetlocks, hocks and stifles. According to their radiographic status (OC negative vs. OC positive), Richards growth function was adjusted to BW and WH data. Instantaneous BW and WH growth rates (BW IADG and WH IADG) were calculated for each foal, from the resolution of the first derivative of growth models for seven age-classes. The presence of radiographic findings compatible with OC at the onset of training was associated with changes in BW and WH growth rates. Positive horses presented higher BW IADG from six to 18 months of age and lower WH IADG before 45 days of age (P<0.001). Speed of sound measurements (SOS), bone markers, growth factors and other metabolic variables change markedly with age (P<0.01). OC positive horses tended to have lower SOS values at the lateral region of McIII, lower IGF-I, and higher insulin and PTH concentrations (P<0.1). This study provides indirect evidence that monitoring foals' growth during the first year of life may be of assistance in managing the occurrence of OC. Further studies with a higher number of animals and a controlled feed intake should be pursued.


Subject(s)
Biomarkers , Bone Development , Cartilage, Articular/growth & development , Cartilage, Articular/metabolism , Animals , Body Weights and Measures , Bone Development/genetics , Female , Horses , Longitudinal Studies , Male
11.
Int J Mol Sci ; 19(12)2018 12 04.
Article in English | MEDLINE | ID: mdl-30518103

ABSTRACT

Cancer is a multi-process disease where different mechanisms exist in parallel to ensure cell survival and constant adaptation to the extracellular environment. To adapt rapidly, cancer cells re-arrange their plasma membranes to sustain proliferation, avoid apoptosis and resist anticancer drugs. In this review, we discuss novel approaches based on the modifications and manipulations that new classes of molecules can exert in the plasma membrane lateral organization and order of cancer cells, affecting growth factor signaling, invasiveness, and drug resistance. Furthermore, we present azurin, an anticancer protein from bacterial origin, as a new approach in the development of therapeutic strategies that target the cell membrane to improve the existing standard therapies.


Subject(s)
Cell Membrane/metabolism , Molecular Targeted Therapy , Neoplasms/pathology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biophysical Phenomena , Cell Membrane/drug effects , Humans , Neoplasms/metabolism
12.
Cell Cycle ; 17(13): 1649-1666, 2018.
Article in English | MEDLINE | ID: mdl-29963969

ABSTRACT

Membrane lipid rafts are highly ordered microdomains and essential components of plasma membranes. In this work, we demonstrate that azurin uptake by cancer cells is, in part, mediated by caveolin-1 and GM-1, lipid rafts' markers. This recognition is mediated by a surface exposed hydrophobic core displayed by azurin since the substitution of a phenylalanine residue in position 114 facing the hydrophobic cavity by alanine impacts such interactions, debilitating the uptake of azurin by cancer cells. Treating of cancer cells with azurin leads to a sequence of events: alters the lipid raft exposure at plasma membranes, causes a decrease in the plasma membrane order as examined by Laurdan two-photon imaging and leads to a decrease in the levels of caveolin-1. Caveolae, a subset of lipid rafts characterized by the presence of caveolin-1, are gaining increasing recognition as mediators in tumor progression and resistance to standard therapies. We show that azurin inhibits growth of cancer cells expressing caveolin-1, and this inhibition is only partially observed with mutant azurin. Finally, the simultaneous administration of azurin with anticancer therapeutic drugs (paclitaxel and doxorubicin) results in an enhancement in their activity, contrary to the mutated protein.


Subject(s)
Antineoplastic Agents/pharmacology , Azurin/metabolism , Caveolin 1/metabolism , G(M1) Ganglioside/metabolism , Membrane Fluidity , Membrane Microdomains/metabolism , Amino Acid Sequence , Azurin/chemistry , Azurin/genetics , Caveolin 1/chemistry , Cell Line, Tumor , Humans , Mutant Proteins/metabolism , Point Mutation/genetics , Protein Domains
13.
Nat Commun ; 8(1): 922, 2017 10 13.
Article in English | MEDLINE | ID: mdl-29030545

ABSTRACT

Yeast and cancer cells share the unusual characteristic of favoring fermentation of sugar over respiration. We now reveal an evolutionary conserved mechanism linking fermentation to activation of Ras, a major regulator of cell proliferation in yeast and mammalian cells, and prime proto-oncogene product. A yeast mutant (tps1∆) with overactive influx of glucose into glycolysis and hyperaccumulation of Fru1,6bisP, shows hyperactivation of Ras, which causes its glucose growth defect by triggering apoptosis. Fru1,6bisP is a potent activator of Ras in permeabilized yeast cells, likely acting through Cdc25. As in yeast, glucose triggers activation of Ras and its downstream targets MEK and ERK in mammalian cells. Biolayer interferometry measurements show that physiological concentrations of Fru1,6bisP stimulate dissociation of the pure Sos1/H-Ras complex. Thermal shift assay confirms direct binding to Sos1, the mammalian ortholog of Cdc25. Our results suggest that the Warburg effect creates a vicious cycle through Fru1,6bisP activation of Ras, by which enhanced fermentation stimulates oncogenic potency.Yeast and cancer cells both favor sugar fermentation in aerobic conditions. Here the authors describe a conserved mechanism from yeast to mammals where the glycolysis intermediate fructose-1,6-bisphosphate binds Cdc25/Sos1 and couples increased glycolytic flux to increased Ras proto-oncoprotein activity.


Subject(s)
Fructosephosphates/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , ras Proteins/metabolism , Animals , Fermentation , Glucose/metabolism , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Glycolysis , SOS1 Protein/genetics , SOS1 Protein/metabolism , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , ras Proteins/genetics , ras-GRF1/genetics , ras-GRF1/metabolism
14.
Cell Cycle ; 15(11): 1415-24, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27096894

ABSTRACT

In lung cancer, the Epidermal Growth Factor Receptor (EGFR) is one of the main targets for clinical management of this disease. The effectiveness of therapies toward this receptor has already been linked to the expression of integrin receptor subunit ß1 in NSCLC A549 cells. In this work we demonstrate that azurin, an anticancer therapeutic protein originated from bacterial cells, controls the levels of integrin ß1 and its appropriate membrane localization, impairing the intracellular signaling cascades downstream these receptors and the invasiveness of cells. We show evidences that azurin when combined with gefitinib and erlotinib, tyrosine kinase inhibitors which targets specifically the EGFR, enhances the sensitivity of these lung cancer cells to these molecules. The broad effect of azurin at the cell surface level was examined by Atomic Force Microscopy. The Young 's module (E) shows that the stiffness of A549 lung cancer cells decreased with exposure to azurin and also gefitinib, suggesting that the alterations in the membrane properties may be the basis of the broad anticancer activity of this protein. Overall, these results show that azurin may be relevant as an adjuvant to improve the effects of other anticancer agents already in clinical use, to which patients often develop resistance hampering its full therapeutic response.


Subject(s)
Antineoplastic Agents/pharmacology , Azurin/pharmacology , Cell Membrane/drug effects , ErbB Receptors/genetics , Erlotinib Hydrochloride/pharmacology , Integrin beta1/genetics , Quinazolines/pharmacology , A549 Cells , Bacterial Proteins/pharmacology , Cell Adhesion/drug effects , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Elastic Modulus , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Gefitinib , Gene Expression , Humans , Integrin beta1/metabolism , Microscopy, Atomic Force , Surface Properties
15.
Bioengineered ; 5(4): 234-42, 2014.
Article in English | MEDLINE | ID: mdl-24875003

ABSTRACT

Cancer is one of the most deadly diseases worldwide. In the last three decades many efforts have been made focused on understanding how cancer grows and responds to drugs. The dominant drug-development paradigm has been the "one drug, one target." Based on that, the two main targeted therapies developed to combat cancer include the use of tyrosine kinase inhibitors and monoclonal antibodies. Development of drug resistance and side effects represent the major limiting factors for their use in cancer treatment. Nowadays, a new paradigm for cancer drug discovery is emerging wherein multi-targeted approaches gain ground in cancer therapy. Therefore, to overcome resistance to therapy, it is clear that a new generation of drugs is urgently needed. Here, regarding the concept of multi-targeted therapy, we discuss the challenges of using bacterial proteins and peptides as a new generation of effective anti-cancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Bacterial Proteins/chemistry , Bacterial Proteins/pharmacology , Antibodies, Monoclonal , Azurin/pharmacology , Drug Discovery , Drug Resistance, Neoplasm , Protein Conformation , Signal Transduction , Transcriptome
16.
Int J Biochem Cell Biol ; 50: 1-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24509127

ABSTRACT

Azurin is a bacterial protein from Pseudomonas aeruginosa which exerts an inhibitory activity in cancer cells. In P-cadherin-overexpressing models, a bad prognosis marker in breast cancer increasing invasion and other malignant features, azurin decreases the invasion of cancer cells. We performed a microarray analysis to compare the expression profile of azurin treated cells with different P-cadherin expression levels. Azurin up-regulated apoptosis mediated by p53 protein, endocytosis and vesicle-mediated transport. In the contrary, in invasive MCF-7/AZ.Pcad cells, azurin decreased the expression of genes associated with cell surface receptors and signal transduction, as well as biological adhesion. Further, azurin decreased adhesion of cells to proteins from the extracellular matrix (ECM) and altered protein expression of integrins α6, ß4 and ß1 and interfered with the ability of these cells to form mammospheres. Altogether, our results further enlighten the anti-cancer effects mediated by azurin in P-cadherin overexpression breast cancer models.


Subject(s)
Azurin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cadherins/biosynthesis , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Adhesion/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , High-Throughput Screening Assays/methods , Humans , Integrins/biosynthesis , Integrins/genetics , MCF-7 Cells , Oligonucleotide Array Sequence Analysis/methods , Pseudomonas aeruginosa/metabolism , Signal Transduction/drug effects , Transcriptome/drug effects , Tumor Suppressor Protein p53/metabolism
17.
PLoS One ; 8(7): e69023, 2013.
Article in English | MEDLINE | ID: mdl-23894398

ABSTRACT

P-cadherin overexpression occurs in about 30% of all breast carcinomas, being a poor prognostic factor for breast cancer patients. In a cellular background of wild-type E-cadherin, we have previously shown that its expression promotes invasion, motility and migration of breast cancer cells due to the induced secretion of metalloproteases (MMPs) to the extracellular medium and to the concomitant shedding of a pro-invasive soluble form of this protein (sP-cad). Azurin is secreted by Pseudomonas aeruginosa and induces in vitro and in vivo cytotoxicity after its preferential penetration in human cancer cells relative to normal cells. Three different breast cancer cell lines, MCF-7/AZ.Mock, MCF-7/AZ.Pcad and SUM149 were treated with sub-killing doses of azurin. Invasion of these cells was measured using Matrigel Invasion Assays and MTT assays were performed to determine cell viability upon treatment and the effects on cadherins expression was determined by Western blot and Immunofluorescence. Gelatin Zymography was used to determine activity of MMP2 in the conditioned media of azurin treated and untreated cells and the phosphorylation levels of intracellular signaling proteins were determined by Western blot. The invasive phenotype of these breast cancer cells was significantly reduced by azurin. Azurin (50-100 µM) also caused a specific decrease on P-cadherin protein levels from 30-50% in MCF-7/AZ.Pcad and SUM149 breast cancer cell lines, but the levels of E-cadherin remain unaltered. More, the levels of sP-cad and the activity of MMP2 were reduced in the extracellular media of azurin treated cells and we also observed a decrease in the phosphorylation levels of both FAK and Src proteins. Our data show that azurin specifically targets P-cadherin, not E-cadherin, abrogating P-cadherin-mediated invasive effects and signaling. Therefore, azurin could possibly be considered a therapeutic tool to treat poor-prognosis breast carcinomas overexpressing P-cadherin in a wild type E-cadherin context.


Subject(s)
Azurin/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cadherins/genetics , Focal Adhesion Kinase 1/metabolism , Gene Expression , Signal Transduction/drug effects , src-Family Kinases/metabolism , Bacterial Proteins/pharmacology , Breast Neoplasms/pathology , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Female , Humans , MCF-7 Cells , Matrix Metalloproteinase 2/metabolism
18.
Appl Microbiol Biotechnol ; 97(12): 5189-99, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23644748

ABSTRACT

The use of live bacteria in cancer therapies offers exciting possibilities. Nowadays, an increasing number of genetically engineered bacteria are emerging in the field, with applications both in therapy and diagnosis. In parallel, purified bacterial products are also gaining relevance as new classes of bioactive products to treat and prevent cancer growth and metastasis. In the first part of the article, we review the latest findings regarding the use of live bacteria and products as anti-cancer agents, paying special attention to immunotoxins, proteins, and peptides. In particular, we focus on the recent results of using azurin or its derived peptide as anticancer therapeutic agents. In the second part, we discuss the challenges of using metagenomic techniques as a distinctive approach for discovering new anti-cancer agents from bacterial origin.


Subject(s)
Antineoplastic Agents/metabolism , Bacteria/metabolism , Biological Therapy/methods , Metabolic Engineering/methods , Metabolic Networks and Pathways/genetics , Neoplasms/diagnosis , Neoplasms/therapy , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/therapeutic use , Azurin/metabolism , Bacteria/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Biotechnology/methods , Computational Biology , Immunotoxins/metabolism , Metagenomics , Technology, Pharmaceutical/methods
19.
FEMS Microbiol Lett ; 328(1): 61-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22150831

ABSTRACT

Burkholderia cepacia complex (Bcc) bacteria are opportunistic pathogens that cause multiresistant pulmonary infections in patients with cystic fibrosis (CF). In this study, we evaluated the in vitro antimicrobial efficacy of eight unsaturated fatty acids against Burkholderia cenocepacia K56-2, a CF epidemic strain. Docosahexaenoic acid (DHA) was the most active compound. Its action can be either bacteriostatic or bactericidal, depending upon the concentration used. The effect of DHA was also evaluated on two others B. cenocepacia clinical isolates and compared with one representative member of all the 17 Bcc species. To test whether DHA could have a therapeutic potential, we assessed its efficacy using a Galleria mellonella caterpillar model of B. cenocepacia infection. We observed that the treatment of infected larvae with a single dose of DHA (50 mM) caused an increase in the survival rate as well as a reduced bacterial load. Moreover, DHA administration markedly increases the expression profile of the gene encoding the antimicrobial peptide gallerimycin. Our results demonstrate that DHA has in vitro and in vivo antibacterial activity against Bcc microorganisms. These findings provide evidence that DHA may be a useful nutraceutical for the treatment of CF patients with lung infections caused by antibiotic multiresistant Bcc microorganisms.


Subject(s)
Anti-Bacterial Agents/pharmacology , Burkholderia cenocepacia/drug effects , Docosahexaenoic Acids/pharmacology , Fatty Acids, Omega-3/pharmacology , Animals , Anti-Bacterial Agents/therapeutic use , Bacterial Load , Burkholderia Infections/microbiology , Burkholderia cenocepacia/growth & development , Burkholderia cenocepacia/isolation & purification , Burkholderia cenocepacia/physiology , Cystic Fibrosis/complications , Disease Models, Animal , Docosahexaenoic Acids/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Humans , Lepidoptera , Microbial Sensitivity Tests , Microbial Viability/drug effects , Pneumonia, Bacterial/microbiology , Survival Analysis
20.
Recent Pat Anticancer Drug Discov ; 7(1): 31-55, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21906014

ABSTRACT

This review intends to provide a comprehensive coverage of the various patents, published or issued, since 2007 on live or attenuated bacteria as potential anticancer agents, as well as microbial products including toxins, enzymes, antibiotics, various proteins and peptides as well as other small molecular weight products. Below is a list of such published/issued patents and a summary of the main contents of many such patents.


Subject(s)
Antineoplastic Agents/therapeutic use , Bacteria/chemistry , Microbial Viability , Neoplasms/drug therapy , Neoplasms/microbiology , Patents as Topic , Animals , Antibiotic Prophylaxis/methods , Antibiotic Prophylaxis/trends , Bacteria/immunology , Humans , Microbial Viability/immunology , Neoplasms/immunology , Salmonella/chemistry , Salmonella/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...