Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
ACS Omega ; 7(23): 20204-20218, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35721990

ABSTRACT

Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis (Mtb). Despite being considered curable and preventable, the increase of antibiotic resistance is becoming a serious public health problem. Mtb is a pathogen capable of surviving in macrophages, causing long-term latent infection where the mycobacterial serine/threonine protein kinase G (PknG) plays a protective role. Therefore, PknG is an important inhibitory target to prevent Mtb from entering the latency stage. In this study, we use a pharmacophore-based virtual screening and biochemical assays to identify the compound RO9021 (CHEMBL3237561) as a PknG inhibitor. In detail, 1.5 million molecules were screened using a scalable cloud-based setup, identifying 689 candidates, which were further subjected to additional screening employing molecular docking. Molecular docking spotted 62 compounds with estimated binding affinities of -7.54 kcal/mol (s.d. = 0.77 kcal/mol). Finally, 14 compounds were selected for in vitro experiments considering previously reported biological activities and commercial availability. In vitro assays of PknG activity showed that RO9021 inhibits the kinase activity similarly to AX20017, a known inhibitor. The inhibitory effect was found to be dose dependent with a relative IC50 value of 4.4 ± 1.1 µM. Molecular dynamics simulations predicted that the PknG-RO9021 complex is stable along the tested timescale. Altogether, our study indicates that RO9021 is a noteworthy drug candidate for further developing new anti-TB drugs that hold excellent reported pharmacokinetic parameters.

2.
Sci Rep ; 11(1): 15452, 2021 07 29.
Article in English | MEDLINE | ID: mdl-34326429

ABSTRACT

SARS-CoV-2 main protease is a common target for inhibition assays due to its high conservation among coronaviruses. Since flavonoids show antiviral activity, several in silico works have proposed them as potential SARS-CoV-2 main protease inhibitors. Nonetheless, there is reason to doubt certain results given the lack of consideration for flavonoid promiscuity or main protease plasticity, usage of short library sizes, absence of control molecules and/or the limitation of the methodology to a single target site. Here, we report a virtual screening study where dorsilurin E, euchrenone a11, sanggenol O and CHEMBL2171598 are proposed to inhibit main protease through different pathways. Remarkably, novel structural mechanisms were observed after sanggenol O and CHEMBL2171598 bound to experimentally proven allosteric sites. The former drastically affected the active site, while the latter triggered a hinge movement which has been previously reported for an inactive SARS-CoV main protease mutant. The use of a curated database of 4.8 k flavonoids, combining two well-known docking software (AutoDock Vina and AutoDock4.2), molecular dynamics and MMPBSA, guaranteed an adequate analysis and robust interpretation. These criteria can be considered for future screening campaigns against SARS-CoV-2 main protease.


Subject(s)
COVID-19/metabolism , Coronavirus 3C Proteases/antagonists & inhibitors , Flavonoids/pharmacology , Antiviral Agents/pharmacology , Binding Sites , COVID-19/immunology , Coronavirus 3C Proteases/immunology , Coronavirus 3C Proteases/metabolism , Databases, Factual , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Protease Inhibitors/pharmacology , Protein Binding , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , COVID-19 Drug Treatment
3.
Front Microbiol ; 12: 682682, 2021.
Article in English | MEDLINE | ID: mdl-34262544

ABSTRACT

Tetracycline has positively impacted human health as well as the farming and animal industries. Its extensive usage and versatility led to the spread of resistance mechanisms followed by the development of new variants of the antibiotic. Tetracyclines inhibit bacterial growth by impeding the binding of elongator tRNAs to the ribosome. However, a small number of reports indicated that Tetracyclines could also inhibit translation initiation, yet the molecular mechanism remained unknown. Here, we use biochemical and computational methods to study how Oxytetracycline (Otc), Demeclocycline (Dem), and Tigecycline (Tig) affect the translation initiation phase of protein synthesis. Our results show that all three Tetracyclines induce Initiation Factor IF3 to adopt a compact conformation on the 30S ribosomal subunit, similar to that induced by Initiation Factor IF1. This compaction was faster for Tig than Dem or Otc. Furthermore, all three tested tetracyclines affected IF1-bound 30S complexes. The dissociation rate constant of IF1 in early 30S complexes was 14-fold slower for Tig than Dem or Otc. Late 30S initiation complexes (30S pre-IC or IC) exhibited greater IF1 stabilization by Tig than for Dem and Otc. Tig and Otc delayed 50S joining to 30S initiation complexes (30S ICs). Remarkably, the presence of Tig considerably slowed the progression to translation elongation and retained IF1 in the resulting 70S initiation complex (70S IC). Molecular modeling of Tetracyclines bound to the 30S pre-IC and 30S IC indicated that the antibiotics binding site topography fluctuates along the initiation pathway. Mainly, 30S complexes show potential contacts between Dem or Tig with IF1, providing a structural rationale for the enhanced affinity of the antibiotics in the presence of the factor. Altogether, our data indicate that Tetracyclines inhibit translation initiation by allosterically perturbing the IF3 layout on the 30S, retaining IF1 during 70S IC formation, and slowing the transition toward translation elongation. Thus, this study describes a new complementary mechanism by which Tetracyclines may inhibit bacterial protein synthesis.

4.
Parasitol Res ; 119(10): 3503-3515, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32772176

ABSTRACT

Malaria, babesiosis, trypanosomosis, and leishmaniasis are some of the most life-threatening parasites, but the range of drugs to treat them is limited. An effective, safe, and low-cost drug with a large activity spectrum is urgently needed. For this purpose, an aryl amino alcohol derivative called Alsinol was resynthesized, screened in silico, and tested against Plasmodium, Babesia, Trypanosoma, and Leishmania. In silico Alsinol follows the Lipinski and Ghose rules. In vitro it had schizontocidal activity against Plasmodium falciparum and was able to inhibit gametocytogenesis; it was particularly active against late gametocytes. In malaria-infected mice, it showed a dose-dependent activity similar to chloroquine. It demonstrated a similar level of activity to reference compounds against Babesia divergens, and against promastigotes, and amastigotes stages of Leishmania in vitro. It inhibited the in vitro growth of two African animal strains of Trypanosoma but was ineffective in vivo in our experimental conditions. It showed moderate toxicity in J774A1 and Vero cell models. The study demonstrated that Alsinol has a large spectrum of activity and is potentially affordable to produce. Nevertheless, challenges remain in the process of scaling up synthesis, creating a suitable clinical formulation, and determining the safety margin in preclinical models.


Subject(s)
Amino Alcohols/pharmacology , Antiprotozoal Agents/pharmacology , Amino Alcohols/chemical synthesis , Amino Alcohols/chemistry , Animals , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Babesia/drug effects , Babesia/growth & development , Cell Survival/drug effects , Chlorocebus aethiops , Disease Models, Animal , Leishmania/drug effects , Leishmania/growth & development , Life Cycle Stages/drug effects , Mice , Plasmodium/drug effects , Plasmodium/growth & development , Protozoan Infections/drug therapy , Protozoan Infections/parasitology , Treatment Outcome , Trypanosoma/drug effects , Trypanosoma/growth & development , Vero Cells
5.
Malar J ; 18(1): 201, 2019 Jun 19.
Article in English | MEDLINE | ID: mdl-31217011

ABSTRACT

BACKGROUND: The challenge in anti-malarial chemotherapy is based on the emergence of resistance to drugs and the search for medicines against all stages of the life cycle of Plasmodium spp. as a therapeutic target. Nowadays, many molecules with anti-malarial activity are reported. However, few studies about the cellular and molecular mechanisms to understand their mode of action have been explored. Recently, new primaquine-based hybrids as new molecules with potential multi-acting anti-malarial activity were reported and two hybrids of primaquine linked to quinoxaline 1,4-di-N-oxide (PQ-QdNO) were identified as the most active against erythrocytic, exoerythrocytic and sporogonic stages. METHODS: To further understand the anti-malarial mode of action (MA) of these hybrids, hepg2-CD81 were infected with Plasmodium yoelii 17XNL and treated with PQ-QdNO hybrids during 48 h. After were evaluated the production of ROS, the mitochondrial depolarization, the total glutathione content, the DNA damage and proteins related to oxidative stress and death cell. RESULTS: In a preliminary analysis as tissue schizonticidals, these hybrids showed a mode of action dependent on peroxides production, but independent of the activation of transcription factor p53, mitochondrial depolarization and arrest cell cycle. CONCLUSIONS: Primaquine-quinoxaline 1,4-di-N-oxide hybrids exert their antiplasmodial activity in the exoerythrocytic phase by generating high levels of oxidative stress which promotes the increase of total glutathione levels, through oxidation stress sensor protein DJ-1. In addition, the role of HIF1a in the mode of action of quinoxaline 1,4-di-N-oxide is independent of biological activity.


Subject(s)
Antimalarials/pharmacology , Plasmodium yoelii/drug effects , Primaquine/pharmacology , Quinoxalines/pharmacology , Drug Combinations , Erythrocytes/parasitology , Hep G2 Cells , Humans , Sporozoites/drug effects
6.
Eur J Med Chem ; 158: 68-81, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30199706

ABSTRACT

Emergence of drug resistance and targeting all stages of the parasite life cycle are currently the major challenges in antimalarial chemotherapy. Molecular hybridization combining two scaffolds in a single molecule is an innovative strategy for achieving these goals. In this work, a series of novel quinoxaline 1,4-di-N-oxide hybrids containing either chloroquine or primaquine pharmacophores was designed, synthesized and tested against both chloroquine sensitive and multidrug resistant strains of Plasmodium falciparum. Only chloroquine-based compounds exhibited potent blood stage activity with compounds 4b and 4e being the most active and selective hybrids at this parasite stage. Based on their intraerythrocytic activity and selectivity or their chemical nature, seven hybrids were then evaluated against the liver stage of Plasmodium yoelii, Plasmodium berghei and Plasmodium falciparum infections. Compound 4b was the only chloroquine-quinoxaline 1,4-di-N-oxide hybrid with a moderate liver activity, whereas compound 6a and 6b were identified as the most active primaquine-based hybrids against exoerythrocytic stages, displaying enhanced liver activity against P. yoelii and P. berghei, respectively, and better SI values than primaquine. Although both primaquine-quinoxaline 1,4-di-N-oxide hybrids slightly reduced the infection of mosquitoes, they inhibited sporogony of P. berghei and compound 6a showed 92% blocking of transmission. In vivo liver efficacy assays revealed that compound 6a showed causal prophylactic activity affording parasitaemia reduction of up to 95% on day 4. Absence of genotoxicity and in vivo acute toxicity were also determined. These results suggest the approach of primaquine-quinoxaline 1,4-di-N-oxide hybrids as new potential dual-acting antimalarials for further investigation.


Subject(s)
Antimalarials/chemistry , Antimalarials/pharmacology , Chloroquine/analogs & derivatives , Chloroquine/pharmacology , Plasmodium/drug effects , Primaquine/analogs & derivatives , Primaquine/pharmacology , Animals , Antimalarials/therapeutic use , Chloroquine/therapeutic use , Female , Hep G2 Cells , Humans , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria/prevention & control , Mice, Inbred BALB C , Plasmodium/physiology , Primaquine/therapeutic use , Quinoxalines/chemistry , Quinoxalines/pharmacology , Quinoxalines/therapeutic use
7.
Eur J Med Chem ; 152: 489-514, 2018 May 25.
Article in English | MEDLINE | ID: mdl-29754074

ABSTRACT

Design, synthesis, structure-activity relationship, cytotoxicity studies, in silico drug-likeness, genotoxicity screening, and in vivo studies of new 1-aryl-3-substituted propanol derivatives led to the identification of nine compounds with promising in vitro (55, 56, 61, 64, 66, and 70-73) and in vivo (66 and 72) antimalarial profiles against Plasmodium falciparum and Plasmodium berghei. Compounds 55, 56, 61, 64, 66 and 70-73 exhibited potent antiplasmodial activity against chloroquine-resistant strain FCR-3 (IC50s < 0.28 µM), and compounds 55, 56, 64, 70, 71, and 72 showed potent biological activity in chloroquine-sensitive and multidrug-resistant strains (IC50s < 0.7 µM for 3D7, D6, FCR-3 and C235). All of these compounds share appropriate drug-likeness profiles and adequate selectivity indexes (77 < SI < 184) as well as lack genotoxicity. In vivo efficacy tests in a mouse model showed compounds 66 and 72 to be promising candidates as they exhibited significant parasitemia reductions of 96.4% and 80.4%, respectively. Additional studies such as liver stage and sporogony inhibition, target exploration of heat shock protein 90 of P. falciparum, targeted delivery by immunoliposomes, and enantiomer characterization were performed and strongly reinforce the hypothesis of 1-aryl-3-substituted propanol derivatives as promising antimalarial compounds.


Subject(s)
Antimalarials/pharmacology , Drug Delivery Systems , Parasitemia/drug therapy , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Propanols/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Mice , Molecular Structure , Parasitemia/parasitology , Parasitic Sensitivity Tests , Plasmodium berghei/growth & development , Plasmodium falciparum/growth & development , Propanols/chemical synthesis , Propanols/chemistry , Structure-Activity Relationship
8.
Biomaterials ; 145: 178-191, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28869864

ABSTRACT

Most drugs currently entering the clinical pipeline for severe malaria therapeutics are of lipophilic nature, with a relatively poor solubility in plasma and large biodistribution volumes. Low amounts of these compounds do consequently accumulate in circulating Plasmodium-infected red blood cells, exhibiting limited antiparasitic activity. These drawbacks can in principle be satisfactorily dealt with by stably encapsulating drugs in targeted nanocarriers. Here this approach has been adapted for its use in immunocompetent mice infected by the Plasmodium yoelii 17XL lethal strain, selected as a model for human blood infections by Plasmodium falciparum. Using immunoliposomes targeted against a surface protein characteristic of the murine erythroid lineage, the protocol has been applied to two novel antimalarial lipophilic drug candidates, an aminoquinoline and an aminoalcohol. Large encapsulation yields of >90% were obtained using a citrate-buffered pH gradient method and the resulting immunoliposomes reached in vivo erythrocyte targeting and retention efficacies of >80%. In P. yoelii-infected mice, the immunoliposomized aminoquinoline succeeded in decreasing blood parasitemia from severe to uncomplicated malaria parasite densities (i.e. from ≥25% to ca. 5%), whereas the same amount of drug encapsulated in non-targeted liposomes had no significant effect on parasite growth. Pharmacokinetic analysis indicated that this good performance was obtained with a rapid clearance of immunoliposomes from the circulation (blood half-life of ca. 2 h), suggesting a potential for improvement of the proposed model.


Subject(s)
Antibodies, Monoclonal/metabolism , Antimalarials/therapeutic use , Drug Delivery Systems , Erythrocytes/metabolism , Lipids/chemistry , Malaria, Falciparum/drug therapy , Malaria, Falciparum/pathology , Polyethylene Glycols/chemistry , Animals , Antimalarials/blood , Antimalarials/chemistry , Antimalarials/pharmacokinetics , Disease Models, Animal , Humans , Immunocompetence , Inhibitory Concentration 50 , Liposomes , Maximum Tolerated Dose , Mice, Inbred BALB C , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development
9.
PLoS One ; 12(4): e0175646, 2017.
Article in English | MEDLINE | ID: mdl-28410387

ABSTRACT

Parasitic infections are generally diagnosed by professionals trained to recognize the morphological characteristics of the eggs in microscopic images of fecal smears. However, this laboratory diagnosis requires medical specialists which are lacking in many of the areas where these infections are most prevalent. In response to this public health issue, we developed a software based on pattern recognition analysis from microscopi digital images of fecal smears, capable of automatically recognizing and diagnosing common human intestinal parasites. To this end, we selected 229, 124, 217, and 229 objects from microscopic images of fecal smears positive for Taenia sp., Trichuris trichiura, Diphyllobothrium latum, and Fasciola hepatica, respectively. Representative photographs were selected by a parasitologist. We then implemented our algorithm in the open source program SCILAB. The algorithm processes the image by first converting to gray-scale, then applies a fourteen step filtering process, and produces a skeletonized and tri-colored image. The features extracted fall into two general categories: geometric characteristics and brightness descriptions. Individual characteristics were quantified and evaluated with a logistic regression to model their ability to correctly identify each parasite separately. Subsequently, all algorithms were evaluated for false positive cross reactivity with the other parasites studied, excepting Taenia sp. which shares very few morphological characteristics with the others. The principal result showed that our algorithm reached sensitivities between 99.10%-100% and specificities between 98.13%- 98.38% to detect each parasite separately. We did not find any cross-positivity in the algorithms for the three parasites evaluated. In conclusion, the results demonstrated the capacity of our computer algorithm to automatically recognize and diagnose Taenia sp., Trichuris trichiura, Diphyllobothrium latum, and Fasciola hepatica with a high sensitivity and specificity.


Subject(s)
Algorithms , Helminthiasis/diagnosis , Animals , Diphyllobothriasis/diagnosis , Diphyllobothrium/growth & development , Fasciola hepatica/growth & development , Fascioliasis/diagnosis , Humans , Image Processing, Computer-Assisted , Microscopy , Ovum/pathology , Pattern Recognition, Automated , Sensitivity and Specificity , Taenia/growth & development , Taeniasis/diagnosis , Trichuriasis/diagnosis , Trichuris/growth & development
10.
Bioorg Med Chem Lett ; 27(8): 1820-1825, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28291694

ABSTRACT

We report the design (in silico ADMET criteria), synthesis, cytotoxicity studies (HepG-2 cells), and biological evaluation of 15 hydrazine/hydrazide quinoxaline 1,4-di-N-oxide derivatives against the 3D7 chloroquine sensitive strain and FCR-3 multidrug resistant strain of Plasmodium falciparum and Leishmania infantum (axenic amastigotes). Fourteen of derivatives are novel quinoxaline 1,4-di-N-oxide derivatives. Compounds 18 (3D7 IC50=1.40µM, FCR-3 IC50=2.56µM) and 19 (3D7 IC50=0.24µM, FCR-3 IC50=2.8µM) were identified as the most active against P. falciparum, and they were the least cytotoxic (CC50-values>241µM) and most selective (SI>86). None of the compounds tested against L. infantum were considered to be active. Additionally, the functional role of the hydrazine and hydrazide structures were studied in the quinoxaline 1,4-di-N-oxide system.


Subject(s)
Antiprotozoal Agents/chemistry , Antiprotozoal Agents/pharmacology , Hydrazines/chemistry , Hydrazines/pharmacology , Quinoxalines/chemistry , Quinoxalines/pharmacology , Humans , Leishmania infantum/drug effects , Leishmaniasis, Visceral/drug therapy , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Structure-Activity Relationship
11.
Int J Parasitol Drugs Drug Resist ; 6(3): 184-198, 2016 12.
Article in English | MEDLINE | ID: mdl-27718413

ABSTRACT

Synthesis of new 1-aryl-3-substituted propanol derivatives followed by structure-activity relationship, in silico drug-likeness, cytotoxicity, genotoxicity, in silico metabolism, in silico pharmacophore modeling, and in vivo studies led to the identification of compounds 22 and 23 with significant in vitro antiplasmodial activity against drug sensitive (D6 IC50 ≤ 0.19 µM) and multidrug resistant (FCR-3 IC50 ≤ 0.40 µM and C235 IC50 ≤ 0.28 µM) strains of Plasmodium falciparum. Adequate selectivity index and absence of genotoxicity was also observed. Notably, compound 22 displays excellent parasitemia reduction (98 ± 1%), and complete cure with all treated mice surviving through the entire period with no signs of toxicity. One important factor is the agreement between in vitro potency and in vivo studies. Target exploration was performed; this chemotype series exhibits an alternative antimalarial mechanism.


Subject(s)
Amino Alcohols/isolation & purification , Amino Alcohols/pharmacology , Antimalarials/isolation & purification , Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Amino Alcohols/adverse effects , Amino Alcohols/therapeutic use , Animals , Antimalarials/adverse effects , Antimalarials/therapeutic use , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Inhibitory Concentration 50 , Malaria, Falciparum/drug therapy , Mice , Structure-Activity Relationship , Survival Analysis , Treatment Outcome
12.
Biochimie ; 127: 133-43, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27163845

ABSTRACT

In this study, we cloned, expressed and purified the isopentenyl diphosphate isomerases (IDIs) from two plants, Hevea brasiliensis and Solanum lycopersicum, and compared them to the already well characterized Escherichia coli IDI. Phylogenetic analysis showed high homology between the three enzymes. Their catalytic activity was investigated in vitro with recombinant purified enzymes and in vivo by complementation colorimetric tests. The three enzymes displayed consistent activities both in vitro and in vivo. In term of structure, studied by ATR-FTIR and molecular modeling, it is clear that both plant enzymes are more related to their human homologue than to E. coli IDI. But it is assumed that EcIDI represent the minimalistic part of the catalytic core, as both plant enzymes present a supplementary sequence forming an extra α-helice surrounding the catalytic site that could facilitate the biocatalysis. New potential biotechnological applications may be envisaged.


Subject(s)
Carbon-Carbon Double Bond Isomerases/chemistry , Carbon-Carbon Double Bond Isomerases/metabolism , Escherichia coli/enzymology , Hevea/enzymology , Solanum lycopersicum/enzymology , Amino Acid Sequence , Biocatalysis , Hemiterpenes , Humans , Models, Molecular , Species Specificity
13.
J Mol Model ; 21(11): 292, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26499499

ABSTRACT

The goal of this study was to identify neuraminidase (NA) residue mutants from human influenza AH1N1 using sequences from 1918 to 2012. Multiple alignment studies of complete NA sequences (5732) were performed. Subsequently, the crystallographic structure of the 1918 influenza (PDB ID: 3BEQ-A) was used as a wild-type structure and three-dimensional (3-D) template for homology modeling of the mutated selected NA sequences. The 3-D mutated NAs were refined using molecular dynamics (MD) simulations (50 ns). The refined 3-D models were used to perform docking studies using oseltamivir. Multiple sequence alignment studies showed seven representative mutations (A232V, K262R, V263I, T264V, S367L, S369N, and S369K). MD simulations applied to 3-D NAs showed that each NA had different active-site shapes according to structural surface visualization and docking results. Moreover, Cartesian principal component analyses (cPCA) show structural differences among these NA structures caused by mutations. These theoretical results suggest that the selected mutations that are located outside of the active site of NA could affect oseltamivir recognition and could be associated with resistance to oseltamivir.


Subject(s)
Influenza A Virus, H1N1 Subtype/enzymology , Neuraminidase/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Binding Sites , Ligands , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Point Mutation , Principal Component Analysis , Protein Structure, Tertiary
14.
Immunol Res ; 56(1): 44-60, 2013 May.
Article in English | MEDLINE | ID: mdl-23371837

ABSTRACT

The neuraminidase (NA) epitope from the Mexican AH1N1 influenza virus was identified by using sequences registered at the GenBank during the peak of a pandemic (from April 2009 to October 2010). First, NA protein sequences were submitted for multiple alignment analysis, and their three-dimensional models (3-D) were then built by using homology modeling. The most common sequence (denominated wild-type) and its mutants were submitted to linear and nonlinear epitope predictors, which included the major histocompatibility complex type II (MHC II) and B-cell peptides. The epitope prediction was in accordance with evolutionary behavior and some protein structural properties. The latter included a low NA mutation rate, NA 3-D surface exposure, and the presence of high hindrance side chain residues. After selecting the epitope, docking studies and molecular dynamics (MD) simulations were used to explore interactions between the epitope and MHC II. Afterward, several experimental assays were performed to validate the theoretical study by using antibodies from humans (infected by pandemic H1N1) and rabbits (epitope vaccination). The results show 119 complete sequences that were grouped into 28 protein sequences according to their identity (one wild-type and 27 representative mutants (1-5 mutations)). The predictors yielded several epitopes, with the best fit being the one located in the C-terminal region. Theoretical methods demonstrated that the selected epitope reached the P4, P6, P7, and P9 pockets of MHC II, whereas the experimental evidence indicates that the epitope is recognized by human antibodies and also by rabbit antibodies immunized with the peptide.


Subject(s)
Epitope Mapping/methods , Epitopes, B-Lymphocyte/metabolism , Host-Pathogen Interactions/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Neuraminidase/metabolism , Orthomyxoviridae Infections/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/metabolism , Computational Biology , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Influenza, Human/diagnosis , Mexico , Models, Animal , Molecular Sequence Data , Mutation/genetics , Neuraminidase/genetics , Neuraminidase/immunology , Protein Binding , Protein Conformation , Rabbits , Vaccination
15.
Biopolymers ; 99(1): 10-21, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23097226

ABSTRACT

The recent occurrence of 2009 influenza A (H1N1) pandemic as well as others has raised concern of a far more dangerous outcome should this virus becomes resistant to current drug therapies. The number of clinical cases that are resistant to oseltamivir (Tamiflu®) is larger than the limited number of neuraminidase (NA) mutations (H275Y, N295S, and I223R) that have been identified at the active site and that are associated to oseltamivir resistance. In this study, we have performed a comparative analysis between a set of NAs that have the most representative mutations located outside the active site. The recently crystallized NA-oseltamivir complex (PDB ID: 3NSS) was used as a wild-type structure. After selecting the target NA sequences, their three-dimensional (3D) structure was built using 3NSS as a template by homology modeling. The 3D NA models were refined by molecular dynamics (MD) simulations. The refined models were used to perform a docking study, using oseltamivir as a ligand. Furthermore, the docking results were refined by free-energy analysis using the MM-PBSA method. The analysis of the MD simulation results showed that the NA models reached convergence during the first 10 ns. Visual inspection and structural measures showed that the mutated NA active sites show structural variations. The docking and MM-PBSA results from the complexes showed different binding modes and free energy values. These results suggest that distant mutations located outside the active site of NA affect its structure and could be considered to be a new source of resistance to oseltamivir, which agrees with reports in the clinical literature.


Subject(s)
Influenza A Virus, H1N1 Subtype/enzymology , Influenza, Human/virology , Mutation , Neuraminidase/chemistry , Neuraminidase/genetics , Amino Acid Sequence , Binding Sites/genetics , Humans , Molecular Dynamics Simulation , Neuraminidase/antagonists & inhibitors , Oseltamivir/chemistry , Sequence Alignment
16.
Mem Inst Oswaldo Cruz ; 107(6): 820-3, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22990975

ABSTRACT

The effectiveness of methylene blue (MB) combined with pyrimethamine (PYR), chloroquine (CQ) or quinine (Q) was examined in a classical four-day suppressive test against a causative agent of rodent malaria, Plasmodium berghei. A marked potentiation was observed when MB was administered at a non-curative dose of 15 mg/kg/day in combination with PYR (0.19 mg/kg/day) or Q (25 mg/kg/day). No synergy was found between MB (15 mg/Kg) and CQ (0.75 mg/Kg). Our results suggest that the combination of MB with PYR or Q may improve the efficacy of these currently used antimalarial drugs.


Subject(s)
Antimalarials/therapeutic use , Enzyme Inhibitors/therapeutic use , Malaria/drug therapy , Methylene Blue/therapeutic use , Animals , Chloroquine/therapeutic use , Disease Models, Animal , Drug Therapy, Combination/methods , Male , Mice , Plasmodium berghei/drug effects , Pyrimethamine/therapeutic use , Quinine/therapeutic use
17.
Mem. Inst. Oswaldo Cruz ; 107(6): 820-823, set. 2012. ilus, tab
Article in English | LILACS | ID: lil-649501

ABSTRACT

The effectiveness of methylene blue (MB) combined with pyrimethamine (PYR), chloroquine (CQ) or quinine (Q) was examined in a classical four-day suppressive test against a causative agent of rodent malaria, Plasmodium berghei. A marked potentiation was observed when MB was administered at a non-curative dose of 15 mg/kg/day in combination with PYR (0.19 mg/kg/day) or Q (25 mg/kg/day). No synergy was found between MB (15 mg/Kg) and CQ (0.75 mg/Kg). Our results suggest that the combination of MB with PYR or Q may improve the efficacy of these currently used antimalarial drugs.


Subject(s)
Animals , Male , Mice , Antimalarials/therapeutic use , Enzyme Inhibitors/therapeutic use , Malaria/drug therapy , Methylene Blue/therapeutic use , Chloroquine/therapeutic use , Disease Models, Animal , Drug Therapy, Combination/methods , Plasmodium berghei/drug effects , Pyrimethamine/therapeutic use , Quinine/therapeutic use
18.
Bioinformation ; 6(9): 335-9, 2011.
Article in English | MEDLINE | ID: mdl-21814390

ABSTRACT

The pncA gene codes the pyrazinamidase of Mycobacterium tuberculosis, which converts pyrazinamide to ammonia and pyrazinoic-acid, the active antituberculous compound. Pyrazinamidase mutations are associated to pyrazinamide-resistant phenotype, however how mutations affect the structure of the pyrazinamidase, and how structural changes affect the enzymatic function and the level of pyrazinamide-resistance is unknown. The structures of mutated pyrazinamidases from twelve Mycobacterium tuberculosis strains and the pyrazinamide-susceptible H37Rv reference strain were modelled using homology modelling and single amino acid replacement. Physical-chemical and structural parameters of each pyrazinamidase were calculated. These parameters were: The change of electrical charge of the mutated amino acid, the change of volume of the mutated amino acid, the change of a special amino acid, the distance of the mutated amino acid to the active site, the distance of the mutated amino acid to the metal-coordination site, and the orientation of the side-chain of the mutated amino acid. The variability of the enzymatic activity of the recombinant pyrazinamidases, and the microbiological susceptibility to pyrazinamide determined by BACTEC 460TB, were modelled in multiple linear regressions. Physical-chemical and structural parameters of the mutated pyrazinamidases were tested as predictors. Structural and physical-chemical variations of the pyrazinamidase explained 75% of the variability of the enzymatic activity, 87% of the variability of the kinetic constant and 40% of the variability of the pyrazinamide-resistance level. Based on computer models of mutated pyrazinamidases, the structural parameters explained a high variability of the enzymatic function, and to a lesser extent the resistance level.

19.
Bioorg Med Chem Lett ; 21(15): 4498-502, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21724395

ABSTRACT

Continuing with our efforts to identify new active compounds against malaria and leishmaniasis, 14 new 3-amino-1,4-di-N-oxide quinoxaline-2-carbonitrile derivatives were synthesized and evaluated for their in vitro antimalarial and antileishmanial activity against Plasmodium falciparum Colombian FCR-3 strain and Leishmania amazonensis strain MHOM/BR/76/LTB-012A. Further computational studies were carried out in order to analyze graphic SAR and ADME properties. The results obtained indicate that compounds with one halogenous group substituted in position 6 and 7 provide an efficient approach for further development of antimalarial and antileishmanial agents. In addition, interesting ADME properties were found.


Subject(s)
Antimalarials/chemistry , Leishmania mexicana/drug effects , Quinoxalines/chemistry , Salicylamides/chemistry , Sulfonamides/chemistry , Trypanocidal Agents/chemistry , Animals , Antimalarials/pharmacokinetics , Antimalarials/toxicity , Macrophages/drug effects , Macrophages/immunology , Mice , Plasmodium falciparum/drug effects , Salicylamides/pharmacokinetics , Salicylamides/toxicity , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity , Trypanocidal Agents/pharmacokinetics , Trypanocidal Agents/toxicity
20.
Bioinformation ; 6(7): 271-4, 2011.
Article in English | MEDLINE | ID: mdl-21738328

ABSTRACT

Cysticercosis is a public health problem in several developing countries. The oncosphere protein TSOL18 is the most immunogenic and protective antigen ever reported against porcine cysticercosis, although no specific epitope has been identified to account for these properties. Recent evidence suggests that protection might be associated with conformational epitopes. Linear epitopes from TSOL18 were computationally predicted and evaluated for immunogenicity and protection against porcine cysticercosis. A synthetic peptide was designed based on predicted linear B cell and T cell epitopes that are exposed on the surface of the theoretically modeled structure of TSOL18. Three surface epitopes from TSOL18 were predicted as immunogenic. A peptide comprising a linear arrangement of these epitopes was chemically synthesized. The capacity of the synthetic peptide to protect pigs against an oral challenge with Taenia solium proglottids was tested in a vaccine trial. The synthetic peptide was able to produce IgG antibodies in pigs and was associated to a reduction of the number of cysts, although was not able to provide complete protection, defined as the complete absence of cysts in necropsy. This study demonstrated that B cell and T cell predicted epitopes from TSOL18 were not able to completely protect pigs against an oral challenge with Taenia solium proglottids. Therefore, other linear epitopes or eventually conformational epitopes may be responsible for the protection conferred by TSOL18.

SELECTION OF CITATIONS
SEARCH DETAIL
...