Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Toxicol In Vitro ; 29(7): 1537-44, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26079942

ABSTRACT

DIG-MSK (demycarosil-3D-ß-D-digitoxosyl mithramycin SK; EC-8042), a novel analogue of mithramycin A, induced autophagy in HCT116 human colon carcinoma and, to a lesser extent, in A2780 human ovarian carcinoma cell lines, which was followed by apoptosis and/or necrotic cell death in a time-dependent way. The effects of DIG-MSK included changes in the expression of a set of genes involved in autophagy, the progression of cells through the different phases of cell cycle, and their halting at the checkpoints. Cells treated with the glucose analogue 2-DG (2-deoxy-D-glucose), which induces autophagy because it impairs cell metabolism, or co-treated with 2-DG plus DIG-MSK, also showed altered gene expression and autophagy. In A2780 cells, some genes involved in autophagy were down-regulated by the different treatments, yet the levels of the proteins they encode could be enough to ensure autophagic flux. In HCT116 cells, up-regulation of several pro-autophagic genes resulted in strong autophagic response. Acidic cell organelles and autophagic flux were more evident in HCT116 than in A2780 cells. DIG-MSK was still cytotoxic in cells that underwent autophagy induced by 2-DG. Therefore, we verified that autophagy resulting from a stress response did not protect cells against DIG-MSK, but, instead, autophagy promoted by either 2-DG or the novel mithralogue can enhance the antitumour activity, which depended on the cell type.


Subject(s)
Antineoplastic Agents/pharmacology , Deoxyglucose/pharmacology , Plicamycin/analogs & derivatives , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ovarian Neoplasms/genetics , Plicamycin/pharmacology
2.
Pharmacol Ther ; 152: 111-24, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25960131

ABSTRACT

Sp1 (specificity protein 1) is a well-known member of a family of transcription factors that also includes Sp2, Sp3 and Sp4, which are implicated in an ample variety of essential biological processes and have been proven important in cell growth, differentiation, apoptosis and carcinogenesis. Sp1 activates the transcription of many cellular genes that contain putative CG-rich Sp-binding sites in their promoters. Sp1 and Sp3 proteins bind to similar, if not the same, DNA tracts and compete for binding, thus they can enhance or repress gene expression. Evidences exist that the Sp-family of proteins regulates the expression of genes that play pivotal roles in cell proliferation and metastasis of various tumors. In patients with a variety of cancers, high levels of Sp1 protein are considered a negative prognostic factor. A plethora of compounds can interfere with the trans-activating activities of Sp1 and other Sp proteins on gene expression. Several pathways are involved in the down-regulation of Sp proteins by compounds with different mechanisms of action, which include not only the direct interference with the binding of Sp proteins to their putative DNA binding sites, but also promoting the degradation of Sp protein factors. Down-regulation of Sp transcription factors and Sp1-regulated genes is drug-dependent and it is determined by the cell context. The acknowledgment that several of those compounds are safe enough might accelerate their introduction into clinical usage in patients with tumors that over-express Sp1.


Subject(s)
Drug Delivery Systems/methods , Gene Targeting/methods , Neoplasms/metabolism , Neoplasms/therapy , Sp1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , Binding Sites/physiology , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/genetics
3.
J Cell Mol Med ; 19(4): 786-98, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25689150

ABSTRACT

Treatment of p53-deficient PC-3 human prostate carcinoma cells with nanomolar concentrations of bis-anthracycline WP631 induced changes in gene expression, which resulted in G2/M cell cycle arrest, autophagy and cell death. The presence of 2-deoxy-D-glucose (2-DG), which induces metabolic stress and autophagy, enhanced the antiproliferative effects of WP631. Changes induced by WP631, 2-DG, or co-treatments with both compounds, in the expression of a variety of genes involved in autophagy and apoptosis were quantified by real-time PCR. They were consistent with a raise in autophagy followed by cell death. Some cells dying from G2/M phase showed features of necrosis like early changes in membrane permeability, while others were dying by apoptosis that occurred in presence of little caspase-3 activity. Our results indicate that WP631 is not only an antiproliferative agent acting on gene transcription, but it can also induce autophagy regardless of the presence of other pro-autophagy stimuli. The development of autophagy seemed to improve the cytotoxicity of WP631 in PC-3 cells. Our results indicate that autophagy would enhance the activity of DNA-binding drugs like WP631 that are potent inhibitors of gene transcription.


Subject(s)
Autophagy/drug effects , Daunorubicin/analogs & derivatives , Tumor Suppressor Protein p53/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/genetics , Beclin-1 , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Daunorubicin/pharmacology , Deoxyglucose/pharmacology , Dose-Response Relationship, Drug , Drug Synergism , Flow Cytometry , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression/drug effects , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sequestosome-1 Protein , Time Factors
4.
PLoS One ; 9(8): e105323, 2014.
Article in English | MEDLINE | ID: mdl-25122166

ABSTRACT

Fasciolosis is considered the most widespread trematode disease affecting grazing animals around the world; it is currently recognised by the World Health Organisation as an emergent human pathogen. Triclabendazole is still the most effective drug against this disease; however, resistant strains have appeared and developing an effective vaccine against this disease has increasingly become a priority. Several bioinformatics tools were here used for predicting B- and T-cell epitopes according to the available data for Fasciola hepatica protein amino acid sequences. BALB/c mice were immunised with the synthetic peptides by using the ADAD vaccination system and several immune response parameters were measured (antibody titres, cytokine levels, T-cell populations) to evaluate their ability to elicit an immune response. Based on the immunogenicity results so obtained, seven peptides were selected to assess their protection-inducing ability against experimental infection with F. hepatica metacercariae. Twenty-four B- or T-epitope-containing peptides were predicted and chemically synthesised. Immunisation of mice with peptides so-called B1, B2, B5, B6, T14, T15 and T16 induced high levels of total IgG, IgG1 and IgG2a (p<0.05) and a mixed Th1/Th2/Th17/Treg immune response, according to IFN-γ, IL-4, IL-17 and IL-10 levels, accompanied by increased CD62L+ T-cell populations. A high level of protection was obtained in mice vaccinated with peptides B2, B5, B6 and T15 formulated in the ADAD vaccination system with the AA0029 immunomodulator. The bioinformatics approach used in the present study led to the identification of seven peptides as vaccine candidates against the infection caused by Fasciola hepatica (a liver-fluke trematode). However, vaccine efficacy must be evaluated in other host species, including those having veterinary importance.


Subject(s)
Epitopes, B-Lymphocyte/immunology , Epitopes, T-Lymphocyte/immunology , Fasciola hepatica/chemistry , Fasciola hepatica/immunology , Fascioliasis/immunology , Peptides/immunology , Amino Acid Sequence , Animals , Antibodies, Helminth/blood , Antibodies, Helminth/immunology , Cluster Analysis , Cytokines/blood , Cytokines/genetics , Disease Models, Animal , Epitopes, B-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/chemistry , Fascioliasis/genetics , Fascioliasis/metabolism , Fascioliasis/mortality , Fascioliasis/parasitology , Fascioliasis/prevention & control , Female , Gene Expression Profiling , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice , Peptides/chemical synthesis , Protozoan Vaccines/immunology
5.
PLoS One ; 9(8): e104687, 2014.
Article in English | MEDLINE | ID: mdl-25110883

ABSTRACT

Ovarian cancer has a poor prognosis due to intrinsic or acquired resistance to some cytotoxic drugs, raising the interest in new DNA-binding agents such as mithramycin analogues as potential chemotherapeutic agents in gynecological cancer. Using a genome-wide approach, we have analyzed gene expression in A2780 human ovarian carcinoma cells treated with the novel mithramycin analogue DIG-MSK (demycarosyl-3D-ß-D-digitoxosyl-mithramycin SK) that binds to C+G-rich DNA sequences. Nanomolar concentrations of DIG-MSK abrogated the expression of genes involved in a variety of cell processes including transcription regulation and tumor development, which resulted in cell death. Some of those genes have been associated with cell proliferation and poor prognosis in ovarian cancer. Sp1 transcription factor regulated most of the genes that were down-regulated by the drug, as well as the up-regulation of other genes mainly involved in response to cell stress. The effect of DIG-MSK in the control of gene expression by other transcription factors was also explored. Some of them, such as CREB, E2F and EGR1, also recognize C/G-rich regions in gene promoters, which encompass potential DIG-MSK binding sites. DIG-MSK affected several biological processes and molecular functions related to transcription and its cellular regulation in A2780 cells, including transcription factor activity. This new compound might be a promising drug for the treatment of ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Genomics , Ovarian Neoplasms/pathology , Plicamycin/analogs & derivatives , Transcription, Genetic/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Ontology , Gene Regulatory Networks/drug effects , Genome, Human/drug effects , Genome, Human/genetics , Humans , Plicamycin/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism
6.
Chem Biol Interact ; 219: 123-32, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24907531

ABSTRACT

DIG-MSK (demycarosyl-3D-ß-D-digitoxosyl-mithramycin SK) is a recently isolated compound of the mithramycin family of antitumor antibiotics, which includes mithramycin A (MTA) and mithramycin SK (MSK). Here, we present evidence that the binding of DIG-MSK to DNA shares the general features of other mithramycins such as the preference for C/G-rich tracts, but there are some differences in the strength of binding and the DNA sequence preferentially recognized by DIG-MSK. We aimed at gaining further insights into the DIG-MSK mechanism of action by direct comparison with the effects of the parental MTA. Similar to MTA, MSK and DIG-MSK accumulated rapidly in A2780, IGROV1 and OVCAR3 human ovarian cancer cell lines, and DIG-MSK was a potent inhibitor of both basal and induced expression of an Sp1-driven luciferase vector. This inhibitory activity was confirmed for the endogenous Sp1 gene and a set of Sp-responsive genes, and compared to that of MTA and MSK. Furthermore, DIG-MSK was stronger than MTA as inhibitor of Sp3-driven transcription and endogenous Sp3 gene expression. Differences in the effects of MTA, MSK and DIG-MSK on gene expression may have a large influence on their biological activities.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Gene Expression Regulation, Neoplastic/physiology , Ovarian Neoplasms/drug therapy , Plicamycin/analogs & derivatives , Sp1 Transcription Factor/physiology , Transcription, Genetic/physiology , Antibiotics, Antineoplastic/therapeutic use , Binding Sites/physiology , Cell Line, Tumor , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/physiology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Kinetics , Plicamycin/pharmacology , Plicamycin/therapeutic use , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction , Sp1 Transcription Factor/genetics , Spectrometry, Fluorescence
7.
Biochem Pharmacol ; 84(9): 1133-42, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22981341

ABSTRACT

The effects of mithramycin SK (MSK) and demycarosyl-3D-ß-D-digitoxosyl-mithramycin SK (DIG-MSK; EC-8042), two novel analogs of the antitumor antibiotic mithramycin A, on gene transcription were examined in human HCT116 colon carcinoma cells by quantitative real-time PCR of 89 genes mainly involved in cell cycle control. Each one of the analogs down-regulated a different set of genes, while only five genes were down-regulated by both compounds. Moreover, other genes were significantly up-regulated, among them p21(WAF1)/CDKN1A which is involved in halting cells at the G1 and G2/M checkpoints. These results are rationalized in terms of MSK or DIG-MSK competition with various transcription factors for binding to consensus C/G-rich tracts encompassed in gene promoters. Changes in cell cycle distribution and protein levels after treatment with every analog were consistent with changes observed in gene expression.


Subject(s)
Antineoplastic Agents/pharmacology , Genes, cdc , Plicamycin/analogs & derivatives , Apoptosis , Binding Sites , G1 Phase Cell Cycle Checkpoints , G2 Phase Cell Cycle Checkpoints , HCT116 Cells , Humans , Necrosis , Plicamycin/pharmacology , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic/drug effects
8.
BMC Struct Biol ; 11: 16, 2011 Mar 28.
Article in English | MEDLINE | ID: mdl-21443791

ABSTRACT

BACKGROUND: The TlyA protein has a controversial function as a virulence factor in Mycobacterium tuberculosis (M. tuberculosis). At present, its dual activity as hemolysin and RNA methyltransferase in M. tuberculosis has been indirectly proposed based on in vitro results. There is no evidence however for TlyA relevance in the survival of tubercle bacilli inside host cells or whether both activities are functionally linked. A thorough analysis of structure prediction for this mycobacterial protein in this study shows the need for reevaluating TlyA's function in virulence. RESULTS: Bioinformatics analysis of TlyA identified a ribosomal protein binding domain (S4 domain), located between residues 5 and 68 as well as an FtsJ-like methyltranferase domain encompassing residues 62 and 247, all of which have been previously described in translation machinery-associated proteins. Subcellular localization prediction showed that TlyA lacks a signal peptide and its hydrophobicity profile showed no evidence of transmembrane helices. These findings suggested that it may not be attached to the membrane, which is consistent with a cytoplasmic localization. Three-dimensional modeling of TlyA showed a consensus structure, having a common core formed by a six-stranded ß-sheet between two α-helix layers, which is consistent with an RNA methyltransferase structure. Phylogenetic analyses showed high conservation of the tlyA gene among Mycobacterium species. Additionally, the nucleotide substitution rates suggested purifying selection during tlyA gene evolution and the absence of a common ancestor between TlyA proteins and bacterial pore-forming proteins. CONCLUSION: Altogether, our manual in silico curation suggested that TlyA is involved in ribosomal biogenesis and that there is a functional annotation error regarding this protein family in several microbial and plant genomes, including the M. tuberculosis genome.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Models, Molecular , Molecular Sequence Annotation , Mycobacterium tuberculosis , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Computational Biology , DNA Restriction Enzymes/genetics , Evolution, Molecular , Hemolysin Proteins/genetics , Humans , Intracellular Space/metabolism , Mice , Molecular Sequence Data , Mycobacterium tuberculosis/cytology , Mycobacterium tuberculosis/pathogenicity , Operon/genetics , Protein Conformation , Protein Transport , Reproducibility of Results , Research Design , Transcription, Genetic , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism
9.
Peptides ; 31(11): 1987-94, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20654670

ABSTRACT

Rhoptries are specialized secretory organelles found in all members of the genus Plasmodium whose proteins have been considered as promising vaccine candidates due to their involvement in cell invasion and the formation of the parasitophorous vacuole (PV). The Plasmodium falciparum Pf34 protein was recently identified as a rhoptry-neck protein located in detergent-resistant microdomains (DRMs) that is expressed in mature intraerythrocytic parasite stages, but its biological function is still unknown. Receptor-ligand assays carried out in this study found that peptides 36,051 ((101)DKKFSESLKAHMDHLKILNN(120)Y), 36,053 ((141)KKYIIKEIQNNKYLNKEKKS(160)), 36,055 ((181)WLESVNNIEEKSNILKNIKS(200)Y) and 36,056 ((201)QLLNNIASLNHTLSEEIKNI(220)Y), located in the central portion of Pf34, were found to establish protease-sensitive interactions of high affinity and specificity with receptors on the surface of red blood cell (RBCs). In vitro assays showed that Pf34 high activity binding peptides (HABPs) inhibit invasion of RBCs by P. falciparum merozoites, therefore suggesting that Pf34 could act as an adhesin during invasion and supporting the inclusion of Pf34 HABPs in further studies to develop antimalarial control methods.


Subject(s)
Erythrocytes/metabolism , Erythrocytes/parasitology , Membrane Glycoproteins/pharmacology , Protozoan Proteins/pharmacology , Amino Acid Sequence , Animals , Erythrocytes/drug effects , Humans , Plasmodium falciparum/chemistry
10.
PLoS Comput Biol ; 6(6): e1000824, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585611

ABSTRACT

The mycobacterial cell envelope has been implicated in the pathogenicity of tuberculosis and therefore has been a prime target for the identification and characterization of surface proteins with potential application in drug and vaccine development. In this study, the genome of Mycobacterium tuberculosis H37Rv was screened using Machine Learning tools that included feature-based predictors, general localizers and transmembrane topology predictors to identify proteins that are potentially secreted to the surface of M. tuberculosis, or to the extracellular milieu through different secretory pathways. The subcellular localization of a set of 8 hypothetically secreted/surface candidate proteins was experimentally assessed by cellular fractionation and immunoelectron microscopy (IEM) to determine the reliability of the computational methodology proposed here, using 4 secreted/surface proteins with experimental confirmation as positive controls and 2 cytoplasmic proteins as negative controls. Subcellular fractionation and IEM studies provided evidence that the candidate proteins Rv0403c, Rv3630, Rv1022, Rv0835, Rv0361 and Rv0178 are secreted either to the mycobacterial surface or to the extracellular milieu. Surface localization was also confirmed for the positive controls, whereas negative controls were located on the cytoplasm. Based on statistical learning methods, we obtained computational subcellular localization predictions that were experimentally assessed and allowed us to construct a computational protocol with experimental support that allowed us to identify a new set of secreted/surface proteins as potential vaccine candidates.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Computational Biology/methods , Mycobacterium tuberculosis/metabolism , Animals , Antibodies, Bacterial/chemistry , Antibodies, Bacterial/metabolism , Artificial Intelligence , Bacterial Outer Membrane Proteins/chemistry , Cell Fractionation , Electrophoresis, Polyacrylamide Gel , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Escherichia coli/metabolism , Immunoblotting , Microscopy, Immunoelectron , Models, Statistical , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/chemistry , Peptides/immunology , Peptides/metabolism , Rabbits , Sonication , Subcellular Fractions/metabolism
11.
J Med Chem ; 53(10): 3907-18, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20411955

ABSTRACT

Detergent resistant membranes (DRMs) of Plasmodium falciparum merozoites contain a large number of glycosylphosphatidylinositol (GPI)-anchored proteins that have been implicated in interactions between merozoites and red blood cells (RBCs). In this study, two cysteine-rich proteins anchored by GPI to merozoite DRMs (Pf92 and Pf113) were studied with the aim of identifying regions actively involved in RBC invasion. By means of binding assays, high-activity binding peptides (HABPs) with a large number of binding sites per RBC were identified in Pf92 and Pf113. The nature of the RBC surface receptors for these HABPs was explored using enzyme-treated RBCs and cross-linking assays. Invasion inhibition and immunofluorescence localization studies suggest that Pf92 and Pf113 are involved in RBC invasion and that their adhesion to RBCs is mediated by such HABPs. Additionally, polymorphism and circular dichroism studies support their inclusion in further studies to design components of an antimalarial vaccine.


Subject(s)
Erythrocytes/metabolism , Membrane Proteins/metabolism , Merozoites/physiology , Peptides/metabolism , Plasmodium falciparum/physiology , Protozoan Proteins/metabolism , Animals , Binding Sites , Chymotrypsin/chemistry , Circular Dichroism , Cross-Linking Reagents/chemistry , Cysteine/metabolism , Detergents/pharmacology , Erythrocytes/chemistry , Erythrocytes/parasitology , Glycosylphosphatidylinositols/metabolism , Host-Parasite Interactions , Immune Sera , Membrane Proteins/genetics , Models, Molecular , Neuraminidase/chemistry , Peptides/genetics , Peptides/immunology , Polymorphism, Genetic , Protein Binding , Protein Structure, Secondary , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Rabbits , Succinimides , Trypsin/chemistry
12.
Vaccine ; 28(14): 2653-63, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20085836

ABSTRACT

In this study, we synthesized the complete sequence of the CLAG-9 protein as 67 20-mer-long non-overlapped peptides and assessed their ability to bind to erythrocytes in receptor-ligand assays. Twenty CLAG-9 peptides were found to have specific high-affinity binding ability to erythrocytes (thereby named as HABPs), with nanomolar dissociation constants. CLAG-9 HABPs interacted with different erythrocyte surface receptors having apparent molecular weights of 85, 63 and 34 kDa. CLAG-9 HABPs binding was also affected by pre-treatment of RBCs with enzymes and inhibited erythrocyte invasion in vitro by up to 72% at 200 microM. These results suggest that some protein fragments of CLAG-9 may be part of the molecular machinery used by malaria parasites to invade erythrocytes, hence supporting their study as possible vaccine candidates.


Subject(s)
Cell Adhesion Molecules/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/parasitology , Plasmodium falciparum/pathogenicity , Protozoan Proteins/metabolism , Virulence Factors/metabolism , Amino Acid Sequence , Animals , Cell Adhesion Molecules/genetics , Humans , Kinetics , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Molecular Sequence Data , Molecular Weight , Peptide Hydrolases/metabolism , Protein Binding , Protozoan Proteins/genetics , Virulence Factors/genetics
13.
J Mol Med (Berl) ; 88(1): 61-74, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19768630

ABSTRACT

The Plasmodium falciparum P0 ribosomal phosphoprotein (PfP0) was identified for the first time by screening a cDNA expression library of P. falciparum parasites with sera from malaria-immune individuals. Due to its localization on the surface of different parasite life-cycle stages (merozoites and gametocytes) and its recognition by invasion-blocking antibodies, PfP0 has been considered a potential malaria-vaccine component. In this study, 16 20-mer-long synthetic peptides spanning the entire PfP0 sequence were evaluated by means of receptor-ligand assays with human red blood cells (RBCs) in order to determine the role played by these peptides in the invasion process. Four RBC high-activity binding peptides (HABPs), located mostly toward the N-terminal region, were identified: HABP 33898 ((1)MAKLSKQQKKQMYIEKLSSL(20)), HABP 33900 ((41)ASVRKSLRGKATILMGKNTRY(60)), HABP 33901 ((61)IRTALKKNLQAVPQIEKLLPY (80)), and HABP 33906 ((161)LIKQGEKVTASSATLLRKFNY(180)). The binding pattern of HABPs 33898 and 33906 to enzyme-treated RBCs suggests receptors of protein nature for these two HABPs, one of which could correspond to a common 58-kDa RBC membrane protein, as indicated by results of cross-linking assays. Both HABPs exhibited high content of alpha-helical features and prevented P. falciparum merozoite invasion to RBCs in vitro by up to 91%. The invasion-blocking ability reported here for these PfP0 HABPs supports their inclusion in immunological studies with the aim of assessing their potential as candidates for a vaccine against P. falciparum malaria.


Subject(s)
Erythrocytes/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Ribosomal Proteins/chemistry , Ribosomal Proteins/metabolism , Amino Acid Sequence , Humans , Molecular Sequence Data , Plasmodium falciparum/chemistry , Protein Binding
14.
Biol Chem ; 391(2-3): 207-217, 2010.
Article in English | MEDLINE | ID: mdl-20030583

ABSTRACT

Mycobacterium tuberculosis infection continues to be a major cause of morbidity and mortality throughout the world. The vast complexity of the intracellular pathogen M. tuberculosis and the diverse mechanisms by which it can invade host cells highlight the importance of developing a fully protective vaccine. Our vaccine development strategy consists of including fragments from multiple mycobacterial proteins involved in cell invasion. The aim of this study was to identify high activity binding peptides (HABPs) in the immunogenic protein Rv1980c from M. tuberculosis H37Rv with the ability to inhibit mycobacterial invasion into U937 monocyte-derived macrophages and A549 cells. The presence and transcription of the Rv1980c gene was assessed in members belonging to the M. tuberculosis complex and other nontuberculous mycobacteria by PCR and RT-PCR, respectively. Cell surface localization was confirmed by immuno-electron microscopy. Three peptides binding with high activity to U937 cells and one to A549 cells were identified. HABPs 31100, 31101, and 31107 inhibited invasion of M. tuberculosis into A549 and U937 cells and therefore could be promising candidates for the design of a subunit-based antituberculous vaccine.


Subject(s)
Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Bacterial Vaccines/immunology , Mycobacterium tuberculosis/physiology , Peptide Fragments/metabolism , Vaccines, Synthetic/immunology , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/immunology , Alveolar Epithelial Cells/microbiology , Bacterial Proteins/analysis , Bacterial Proteins/chemistry , Bacterial Vaccines/chemistry , Binding Sites , Blotting, Western , Cells, Cultured , Humans , Models, Molecular , Mycobacterium tuberculosis/genetics , Peptide Fragments/analysis , Peptide Fragments/chemistry , Peptide Fragments/immunology , Transcription, Genetic , Vaccines, Synthetic/chemistry
15.
Vaccine ; 27(49): 6877-86, 2009 Nov 16.
Article in English | MEDLINE | ID: mdl-19755146

ABSTRACT

Severe malaria pathology is directly associated with cytoadherence of infected red blood cells (iRBCs) to healthy RBCs and/or endothelial cells occurring during the intraerythrocytic development of Plasmodium falciparum. We synthesized, as 20-mer long peptides, the members of the ring exported (REX) protein family encoded in chromosome 9, as well as the early transcribed membrane proteins (E-TRAMP) 10.2 and 4, to identify specific RBC binding regions in these proteins. Twelve binding peptides were identified (designated as HABPs): three were identified in REX1, two in REX2, one in REX3, two in REX4 and four in E-TRAMP 10.2. The majority of these HABPs was conserved among different P. falciparum strains, according to sequence analysis. No HABPs were found in E-TRAMP 4. Bindings of HABPs were saturable and sensitive to the enzymatic treatment of RBCs and HABPs had different structural features, according to circular dichroism studies. Our results suggest that the REX and E-TRAMP families participate in relevant interactions with RBC membrane proteins, which highlight these proteins as potential targets for the development of fully effective immunoprophylactic methods.


Subject(s)
Erythrocytes/immunology , Peptides/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Amino Acid Sequence , DNA, Protozoan/genetics , Erythrocyte Membrane/immunology , Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Humans , Malaria Vaccines/immunology , Molecular Sequence Data , Peptides/chemistry , Plasmodium falciparum/genetics , Polymorphism, Genetic , Protein Binding , Protozoan Proteins/chemistry , Sensitivity and Specificity
16.
BMC Bioinformatics ; 10: 134, 2009 May 07.
Article in English | MEDLINE | ID: mdl-19422713

ABSTRACT

BACKGROUND: The computational prediction of mycobacterial proteins' subcellular localization is of key importance for proteome annotation and for the identification of new drug targets and vaccine candidates. Several subcellular localization classifiers have been developed over the past few years, which have comprised both general localization and feature-based classifiers. Here, we have validated the ability of different bioinformatics approaches, through the use of SignalP 2.0, TatP 1.0, LipoP 1.0, Phobius, PA-SUB 2.5, PSORTb v.2.0.4 and Gpos-PLoc, to predict secreted bacterial proteins. These computational tools were compared in terms of sensitivity, specificity and Matthew's correlation coefficient (MCC) using a set of mycobacterial proteins having less than 40% identity, none of which are included in the training data sets of the validated tools and whose subcellular localization have been experimentally confirmed. These proteins belong to the TBpred training data set, a computational tool specifically designed to predict mycobacterial proteins. RESULTS: A final validation set of 272 mycobacterial proteins was obtained from the initial set of 852 mycobacterial proteins. According to the results of the validation metrics, all tools presented specificity above 0.90, while dispersion sensitivity and MCC values were above 0.22. PA-SUB 2.5 presented the highest values; however, these results might be biased due to the methodology used by this tool. PSORTb v.2.0.4 left 56 proteins out of the classification, while Gpos-PLoc left just one protein out. CONCLUSION: Both subcellular localization approaches had high predictive specificity and high recognition of true negatives for the tested data set. Among those tools whose predictions are not based on homology searches against SWISS-PROT, Gpos-PLoc was the general localization tool with the best predictive performance, while SignalP 2.0 was the best tool among the ones using a feature-based approach. Even though PA-SUB 2.5 presented the highest metrics, it should be taken into account that this tool was trained using all proteins reported in SWISS-PROT, which includes the protein set tested in this study, either as a BLAST search or as a training model.


Subject(s)
Bacterial Proteins/analysis , Computational Biology/methods , Mycobacterium/chemistry , Software , Algorithms , Bacterial Proteins/chemistry , Databases, Protein , Mycobacterium/metabolism
17.
Protein Sci ; 17(9): 1494-504, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18556472

ABSTRACT

The identification of proteins present on the surface of Plasmodium falciparum-infected red blood cells as well as of free merozoites has been widely considered as one of the main areas of research in the development of an antimalarial vaccine due to their involvement in the parasite's pathogenesis and invasion mechanisms. Major advances had been accomplished in this area thanks to the analysis of the reported genomic sequence of P. falciparum, allowing for the identification of genes encoding for putative integral membrane proteins. This study reports for the first time the transcription of the MAL8P1.3 gene, which codifies for a 25-kDa integral membrane protein of P. falciparum (FCB-2 strain), namely, Pf25-IMP. Western blot and immunofluorescence assays using goat polyclonal sera indicate that this protein is expressed in erythrocytic asexual blood stages. A highly robust, sensible, and specific receptor-ligand interaction assay allowed identification of two high activity binding peptides (HABPs) derived from Pf25-IMP: 30577 ((41)YKTANENVKLASSLSDRLSR(60)) and 30583 ((161)LNKKTVVRKIAEGLGYTIVF(180)). Both HABPs bound with high affinity to human red blood cells (RBCs), and such binding was susceptible to enzyme treatment with trypsin. A common RBC surface receptor of apparently 48 kDa was found for both HABPs, plus an additional 31-kDa receptor for HABP 30577. HABP 30577 inhibited merozoite invasion in vitro by 73%, while HABP 30583 showed a 59% inhibition at 200 microM concentration. The data suggest a possible role of Pf25-IMP in merozoite invasion to RBCs and support its inclusion in further immunological studies for evaluating its potential as vaccine candidates.


Subject(s)
Erythrocytes/metabolism , Membrane Proteins/metabolism , Merozoites/metabolism , Peptide Fragments/chemistry , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cells, Cultured , Chymotrypsin/pharmacology , Dose-Response Relationship, Drug , Genes, Protozoan , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Merozoites/drug effects , Merozoites/ultrastructure , Molecular Sequence Data , Molecular Weight , Neuraminidase/pharmacology , Peptide Fragments/chemical synthesis , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Protozoan Proteins/pharmacology , Sequence Homology, Amino Acid , Transcription, Genetic , Trypsin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...