Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Med Chem ; 269: 116308, 2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38503166

RESUMO

Plasmodium multi-resistance, including against artemisinin, seriously threatens malaria treatment and control. Hence, new drugs are urgently needed, ideally targeting different parasitic stages, which are not yet targeted by current drugs. The SUB1 protease is involved in both hepatic and blood stages due to its essential role in the egress of parasites from host cells, and, as potential new target, it would meet the above criteria. We report here the synthesis as well as the biological and structural evaluation of substrate-based α-ketoamide SUB1 pseudopeptidic inhibitors encompassing positions P4-P2'. By individually substituting each position of the reference compound 1 (MAM-117, Ac-Ile-Thr-Ala-AlaCO-Asp-Glu (Oall)-NH2), we better characterized the structural determinants for SUB1 binding. We first identified compound 8 with IC50 values of 50 and 570 nM against Pv- and PfSUB1, respectively (about 3.5-fold higher potency compared to 1). Compound 8 inhibited P. falciparum merozoite egress in culture by 37% at 100 µM. By increasing the overall hydrophobicity of the compounds, we could improve the PfSUB1 inhibition level and antiparasitic activity, as shown with compound 40 (IC50 values of 12 and 10 nM against Pv- and PfSUB1, respectively, IC50 value of 23 µM on P. falciparum merozoite egress). We also found that 8 was highly selective towards SUB1 over three mammalian serine peptidases, supporting the promising value of this compound. Finally, several crystal 3D-structures of SUB1-inhibitor complexes, including with 8, were solved at high resolution to decipher the binding mode of these compounds.


Assuntos
Antimaláricos , Malária Falciparum , Malária , Parasitos , Animais , Subtilisina/metabolismo , Sequência de Aminoácidos , Plasmodium falciparum/metabolismo , Peptídeos , Malária Falciparum/parasitologia , Serina Proteases/metabolismo , Relação Estrutura-Atividade , Antimaláricos/farmacologia , Antimaláricos/química , Proteínas de Protozoários , Mamíferos/metabolismo
2.
mBio ; 15(3): e0019824, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38386597

RESUMO

Malaria symptoms are associated with the asexual multiplication of Plasmodium falciparum within human red blood cells (RBCs) and fever peaks coincide with the egress of daughter merozoites following the rupture of the parasitophorous vacuole (PV) and the RBC membranes. Over the last two decades, it has emerged that the release of competent merozoites is tightly regulated by a complex cascade of events, including the unusual multi-step activation mechanism of the pivotal subtilisin-like protease 1 (Sub1) that takes place in three different cellular compartments and remains poorly understood. Following an initial auto-maturation in the endoplasmic reticulum (ER) between its pro- and catalytic domains, the Sub1 prodomain (PD) undergoes further cleavages by the parasite aspartic protease plasmepsin X (PmX) within acidic secretory organelles that ultimately lead to full Sub1 activation upon discharge into the PV. Here, we report the crystal structure of full-length P. falciparum Sub1 (PfS1FL) and demonstrate, through structural, biochemical, and biophysical studies, that the atypical Plasmodium-specific Sub1 PD directly promotes the assembly of inactive enzyme homodimers at acidic pH, whereas Sub1 is primarily monomeric at neutral pH. Our results shed new light into the finely tuned Sub1 spatiotemporal activation during secretion, explaining how PmX processing and full activation of Sub1 can occur in different cellular compartments, and uncover a robust mechanism of pH-dependent subtilisin autoinhibition that plays a key role in P. falciparum merozoites egress from infected host cells.IMPORTANCEMalaria fever spikes are due to the rupture of infected erythrocytes, allowing the egress of Plasmodium sp. merozoites and further parasite propagation. This fleeting tightly regulated event involves a cascade of enzymes, culminating with the complex activation of the subtilisin-like protease 1, Sub1. Differently than other subtilisins, Sub1 activation strictly depends upon the processing by a parasite aspartic protease within acidic merozoite secretory organelles. However, Sub1 biological activity is required in the pH neutral parasitophorous vacuole, to prime effectors involved in the rupture of the vacuole and erythrocytic membranes. Here, we show that the unusual, parasite-specific Sub1 prodomain is directly responsible for its acidic-dependent dimerization and autoinhibition, required for protein secretion, before its full activation at neutral pH in a monomeric form. pH-dependent Sub1 dimerization defines a novel, essential regulatory element involved in the finely tuned spatiotemporal activation of the egress of competent Plasmodium merozoites.


Assuntos
Malária Falciparum , Plasmodium , Animais , Humanos , Subtilisina/metabolismo , Merozoítos/fisiologia , Dimerização , Proteínas de Protozoários/metabolismo , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo , Eritrócitos/parasitologia , Concentração de Íons de Hidrogênio
3.
Acta Crystallogr D Struct Biol ; 79(Pt 8): 721-734, 2023 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-37428845

RESUMO

The constant selection and propagation of multi-resistant Plasmodium sp. parasites require the identification of new antimalarial candidates involved in as-yet untargeted metabolic pathways. Subtilisin-like protease 1 (SUB1) belongs to a new generation of drug targets because it plays a crucial role during egress of the parasite from infected host cells at different stages of its life cycle. SUB1 is characterized by an unusual pro-region that tightly interacts with its cognate catalytic domain, thus precluding 3D structural analysis of enzyme-inhibitor complexes. In the present study, to overcome this limitation, stringent ionic conditions and controlled proteolysis of recombinant full-length P. vivax SUB1 were used to obtain crystals of an active and stable catalytic domain (PvS1Cat) without a pro-region. High-resolution 3D structures of PvS1Cat, alone and in complex with an α-ketoamide substrate-derived inhibitor (MAM-117), showed that, as expected, the catalytic serine of SUB1 formed a covalent bond with the α-keto group of the inhibitor. A network of hydrogen bonds and hydrophobic interactions stabilized the complex, including at the P1' and P2' positions of the inhibitor, although P' residues are usually less important in defining the substrate specificity of subtilisins. Moreover, when associated with a substrate-derived peptidomimetic inhibitor, the catalytic groove of SUB1 underwent significant structural changes, particularly in its S4 pocket. These findings pave the way for future strategies for the design of optimized SUB1-specific inhibitors that may define a novel class of antimalarial candidates.


Assuntos
Antimaláricos , Subtilisina , Plasmodium vivax , Antimaláricos/farmacologia , Antimaláricos/química , Inibidores Enzimáticos/farmacologia , Proteínas de Protozoários/química
4.
J Antimicrob Chemother ; 78(2): 411-417, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36508338

RESUMO

BACKGROUND: In early 2016, in Preah Vihear, Northern Cambodia, artesunate/mefloquine was used to cope with dihydroartemisinin/piperaquine-resistant Plasmodium falciparum parasites. Following this policy, P. falciparum strains harbouring molecular markers associated with artemisinin, piperaquine and mefloquine resistance have emerged. However, the lack of a viable alternative led Cambodia to adopt artesunate/mefloquine countrywide, raising concerns about a surge of triple-resistant P. falciparum strains. OBJECTIVES: To assess the prevalence of triple-resistant parasites after artesunate/mefloquine implementation countrywide in Cambodia and to characterize their phenotype. METHODS: For this multicentric study, 846 samples were collected from 2016 to 2019. Genotyping of molecular markers associated with artemisinin, piperaquine and mefloquine resistance was coupled with phenotypic analyses. RESULTS: Only four triple-resistant P. falciparum isolates (0.47%) were identified during the study period. These parasites combined the pfk13 polymorphism with pfmdr1 amplification, pfpm2 amplification and/or pfcrt mutations. They showed significantly higher tolerance to artemisinin, piperaquine and mefloquine and also to the mefloquine and piperaquine combination. CONCLUSIONS: The use of artesunate/mefloquine countrywide in Cambodia has not led to a massive increase of triple-resistant P. falciparum parasites. However, these parasites circulate in the population, and exhibit clear resistance to piperaquine, mefloquine and their combination in vitro. This study demonstrates that P. falciparum can adapt to more complex drug associations, which should be considered in future therapeutic designs.


Assuntos
Antimaláricos , Artemisininas , Malária Falciparum , Quinolinas , Humanos , Mefloquina/farmacologia , Mefloquina/uso terapêutico , Plasmodium falciparum/genética , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Artesunato , Camboja/epidemiologia , Prevalência , Artemisininas/farmacologia , Artemisininas/uso terapêutico , Quinolinas/farmacologia , Malária Falciparum/tratamento farmacológico , Malária Falciparum/epidemiologia , Malária Falciparum/parasitologia , Resistência a Medicamentos/genética
5.
Lancet Infect Dis ; 17(2): 174-183, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27818097

RESUMO

BACKGROUND: Western Cambodia is the epicentre of Plasmodium falciparum multidrug resistance and is facing high rates of dihydroartemisinin-piperaquine treatment failures. Genetic tools to detect the multidrug-resistant parasites are needed. Artemisinin resistance can be tracked using the K13 molecular marker, but no marker exists for piperaquine resistance. We aimed to identify genetic markers of piperaquine resistance and study their association with dihydroartemisinin-piperaquine treatment failures. METHODS: We obtained blood samples from Cambodian patients infected with P falciparum and treated with dihydroartemisinin-piperaquine. Patients were followed up for 42 days during the years 2009-15. We established in-vitro and ex-vivo susceptibility profiles for a subset using piperaquine survival assays. We determined whole-genome sequences by Illumina paired-reads sequencing, copy number variations by qPCR, RNA concentrations by qRT-PCR, and protein concentrations by immunoblotting. Fisher's exact and non-parametric Wilcoxon rank-sum tests were used to identify significant differences in single-nucleotide polymorphisms or copy number variants, respectively, for differential distribution between piperaquine-resistant and piperaquine-sensitive parasite lines. FINDINGS: Whole-genome exon sequence analysis of 31 culture-adapted parasite lines associated amplification of the plasmepsin 2-plasmepsin 3 gene cluster with in-vitro piperaquine resistance. Ex-vivo piperaquine survival assay profiles of 134 isolates correlated with plasmepsin 2 gene copy number. In 725 patients treated with dihydroartemisinin-piperaquine, multicopy plasmepsin 2 in the sample collected before treatment was associated with an adjusted hazard ratio (aHR) for treatment failure of 20·4 (95% CI 9·1-45·5, p<0·0001). Multicopy plasmepsin 2 predicted dihydroartemisinin-piperaquine failures with 0·94 (95% CI 0·88-0·98) sensitivity and 0·77 (0·74-0·81) specificity. Analysis of samples collected across the country from 2002 to 2015 showed that the geographical and temporal increase of the proportion of multicopy plasmepsin 2 parasites was highly correlated with increasing dihydroartemisinin-piperaquine treatment failure rates (r=0·89 [95% CI 0·77-0·95], p<0·0001, Spearman's coefficient of rank correlation). Dihydroartemisinin-piperaquine efficacy at day 42 fell below 90% when the proportion of multicopy plasmepsin 2 parasites exceeded 22%. INTERPRETATION: Piperaquine resistance in Cambodia is strongly associated with amplification of plasmepsin 2-3, encoding haemoglobin-digesting proteases, regardless of the location. Multicopy plasmepsin 2 constitutes a surrogate molecular marker to track piperaquine resistance. A molecular toolkit combining plasmepsin 2 with K13 and mdr1 monitoring should provide timely information for antimalarial treatment and containment policies. FUNDING: Institut Pasteur in Cambodia, Institut Pasteur Paris, National Institutes of Health, WHO, Agence Nationale de la Recherche, Investissement d'Avenir programme, Laboratoire d'Excellence Integrative "Biology of Emerging Infectious Diseases".


Assuntos
Antimaláricos/uso terapêutico , Artemisininas/uso terapêutico , Biomarcadores/metabolismo , Estudos de Associação Genética , Malária Falciparum/tratamento farmacológico , Quinolinas/uso terapêutico , Ácido Aspártico Endopeptidases , Camboja , Variações do Número de Cópias de DNA/genética , Resistência a Múltiplos Medicamentos , Humanos , Plasmodium falciparum/genética , Plasmodium falciparum/isolamento & purificação , Falha de Tratamento
6.
PLoS One ; 9(10): e109269, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25343504

RESUMO

BACKGROUND: Malaria remains a major global health concern. The development of novel therapeutic strategies is critical to overcome the selection of multiresistant parasites. The subtilisin-like protease (SUB1) involved in the egress of daughter Plasmodium parasites from infected erythrocytes and in their subsequent invasion into fresh erythrocytes has emerged as an interesting new drug target. FINDINGS: Using a computational approach based on homology modeling, protein-protein docking and mutation scoring, we designed protein-based inhibitors of Plasmodium vivax SUB1 (PvSUB1) and experimentally evaluated their inhibitory activity. The small peptidic trypsin inhibitor EETI-II was used as scaffold. We mutated residues at specific positions (P4 and P1) and calculated the change in free-energy of binding with PvSUB1. In agreement with our predictions, we identified a mutant of EETI-II (EETI-II-P4LP1W) with a Ki in the medium micromolar range. CONCLUSIONS: Despite the challenges related to the lack of an experimental structure of PvSUB1, the computational protocol we developed in this study led to the design of protein-based inhibitors of PvSUB1. The approach we describe in this paper, together with other examples, demonstrates the capabilities of computational procedures to accelerate and guide the design of novel proteins with interesting therapeutic applications.


Assuntos
Malária Vivax/metabolismo , Simulação de Acoplamento Molecular , Plasmodium vivax/química , Proteínas de Protozoários/química , Subtilisinas/química , Catálise , Cristalografia por Raios X , Humanos , Malária Vivax/patologia , Plasmodium vivax/metabolismo , Ligação Proteica , Conformação Proteica , Proteínas de Protozoários/genética , Homologia de Sequência de Aminoácidos , Subtilisinas/genética
7.
Nat Commun ; 5: 4833, 2014 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-25204226

RESUMO

The Plasmodium subtilase SUB1 plays a pivotal role during the egress of malaria parasites from host hepatocytes and erythrocytes. Here we report the crystal structure of full-length SUB1 from the human-infecting parasite Plasmodium vivax, revealing a bacterial-like catalytic domain in complex with a Plasmodium-specific prodomain. The latter displays a novel architecture with an amino-terminal insertion that functions as a 'belt', embracing the catalytic domain to further stabilize the quaternary structure of the pre-protease, and undergoes calcium-dependent autoprocessing during subsequent activation. Although dispensable for recombinant enzymatic activity, the SUB1 'belt' could not be deleted in Plasmodium berghei, suggesting an essential role of this domain for parasite development in vivo. The SUB1 structure not only provides a valuable platform to develop new anti-malarial candidates against this promising drug target, but also defines the Plasmodium-specific 'belt' domain as a key calcium-dependent regulator of SUB1 during parasite egress from host cells.


Assuntos
Plasmodium berghei , Plasmodium vivax , Proteínas de Protozoários/metabolismo , Subtilisinas/metabolismo , Sequência de Aminoácidos , Antimaláricos/uso terapêutico , Cristalografia , Humanos , Malária Vivax/tratamento farmacológico , Dados de Sequência Molecular , Terapia de Alvo Molecular , Estrutura Terciária de Proteína
8.
J Biol Chem ; 288(25): 18561-73, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23653352

RESUMO

Widespread drug resistance calls for the urgent development of new antimalarials that target novel steps in the life cycle of Plasmodium falciparum and Plasmodium vivax. The essential subtilisin-like serine protease SUB1 of Plasmodium merozoites plays a dual role in egress from and invasion into host erythrocytes. It belongs to a new generation of attractive drug targets against which specific potent inhibitors are actively searched. We characterize here the P. vivax SUB1 enzyme and show that it displays a typical auto-processing pattern and apical localization in P. vivax merozoites. To search for small PvSUB1 inhibitors, we took advantage of the similarity of SUB1 with bacterial subtilisins and generated P. vivax SUB1 three-dimensional models. The structure-based virtual screening of a large commercial chemical compounds library identified 306 virtual best hits, of which 37 were experimentally confirmed inhibitors and 5 had Ki values of <50 µM for PvSUB1. Interestingly, they belong to different chemical families. The most promising competitive inhibitor of PvSUB1 (compound 2) was equally active on PfSUB1 and displayed anti-P. falciparum and Plasmodium berghei activity in vitro and in vivo, respectively. Compound 2 inhibited the endogenous PfSUB1 as illustrated by the inhibited maturation of its natural substrate PfSERA5 and inhibited parasite egress and subsequent erythrocyte invasion. These data indicate that the strategy of in silico screening of three-dimensional models to select for virtual inhibitors combined with stringent biological validation successfully identified several inhibitors of the PvSUB1 enzyme. The most promising hit proved to be a potent cross-inhibitor of PlasmodiumSUB1, laying the groundwork for the development of a globally active small compound antimalarial.


Assuntos
Plasmodium vivax/enzimologia , Estrutura Terciária de Proteína , Proteínas de Protozoários/química , Serina Proteases/química , Sequência de Aminoácidos , Animais , Antimaláricos/química , Antimaláricos/farmacologia , Sítios de Ligação/genética , Biocatálise/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Feminino , Cinética , Malária/parasitologia , Malária/prevenção & controle , Merozoítos/efeitos dos fármacos , Merozoítos/enzimologia , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/enzimologia , Plasmodium vivax/efeitos dos fármacos , Plasmodium vivax/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Homologia de Sequência de Aminoácidos , Serina Proteases/genética , Serina Proteases/metabolismo , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia , Células Sf9 , Especificidade por Substrato
9.
Methods Mol Biol ; 923: 523-34, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22990802

RESUMO

Drug discovery programs heavily rely on assays adequately monitoring the activity of the drug on the -parasite stage targeted. So far, assays used to screen molecules active against Plasmodium falciparum parasites have mostly been based on measuring growth inhibition of asexual blood stages. We have developed a robust protocol allowing for monitoring parasite egress at the late schizont stage and subsequent erythrocyte invasion. This cytometry-based methodology uses nucleic acid labelling by the dye YOYO-1 and synchronized in vitro culture of P.falciparum exposed to inhibitors during the late phase of the intraerythrocytic cycle and the reinvasion process. This cytometry-based method is quick, accurate and allows for distinguishing egress from reinvasion on thousands of events. The throughput is also increased, as the assay can be scaled up for medium throughput screening for compounds that inhibit either the egress of merozoites or their entry into host erythrocytes.


Assuntos
Antimaláricos/farmacologia , Eritrócitos/parasitologia , Citometria de Fluxo , Merozoítos/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/crescimento & desenvolvimento , Técnicas de Cultura de Células , Eritrócitos/efeitos dos fármacos , Humanos , Merozoítos/metabolismo , Testes de Sensibilidade Parasitária/métodos , Esquizontes/efeitos dos fármacos , Esquizontes/metabolismo
10.
PLoS One ; 6(7): e21812, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21818266

RESUMO

BACKGROUND: Psalmopeotoxin I (PcFK1), a protein of 33 aminoacids derived from the venom of the spider Psalmopoeus Cambridgei, is able to inhibit the growth of Plasmodium falciparum malaria parasites with an IC50 in the low micromolar range. PcFK1 was proposed to act as an ion channel inhibitor, although experimental validation of this mechanism is lacking. The surface loops of PcFK1 have some sequence similarity with the parasite protein sequences cleaved by PfSUB1, a subtilisin-like protease essential for egress of Plasmodium falciparum merozoites and invasion into erythrocytes. As PfSUB1 has emerged as an interesting drug target, we explored the hypothesis that PcFK1 targeted PfSUB1 enzymatic activity. FINDINGS: Molecular modeling and docking calculations showed that one loop could interact with the binding site of PfSUB1. The calculated free energy of binding averaged -5.01 kcal/mol, corresponding to a predicted low-medium micromolar constant of inhibition. PcFK1 inhibited the enzymatic activity of the recombinant PfSUB1 enzyme and the in vitro P. falciparum culture in a range compatible with our bioinformatics analysis. Using contact analysis and free energy decomposition we propose that residues A14 and Q15 are important in the interaction with PfSUB1. CONCLUSIONS: Our computational reverse engineering supported the hypothesis that PcFK1 targeted PfSUB1, and this was confirmed by experimental evidence showing that PcFK1 inhibits PfSUB1 enzymatic activity. This outlines the usefulness of advanced bioinformatics tools to predict the function of a protein structure. The structural features of PcFK1 represent an interesting protein scaffold for future protein engineering.


Assuntos
Peptídeos/farmacologia , Plasmodium falciparum/metabolismo , Engenharia de Proteínas/métodos , Proteínas de Protozoários/antagonistas & inibidores , Genética Reversa/métodos , Venenos de Aranha/farmacologia , Sequência de Aminoácidos , Biocatálise/efeitos dos fármacos , Merozoítos/citologia , Merozoítos/efeitos dos fármacos , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Peptídeos/química , Plasmodium falciparum/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas de Protozoários/química , Proteínas de Protozoários/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Solventes , Venenos de Aranha/química , Subtilisina/metabolismo
11.
J Exp Med ; 206(4): 953-66, 2009 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-19349466

RESUMO

Plasmodium and Toxoplasma are parasites of major medical importance that belong to the Apicomplexa phylum of protozoa. These parasites transform into various stages during their life cycle and express a specific set of proteins at each stage. Although little is yet known of how gene expression is controlled in Apicomplexa, histone modifications, particularly acetylation, are emerging as key regulators of parasite differentiation and stage conversion. We investigated the anti-Apicomplexa effect of FR235222, a histone deacetylase inhibitor (HDACi). We show that FR235222 is active against a variety of Apicomplexa genera, including Plasmodium and Toxoplasma, and is more potent than other HDACi's such as trichostatin A and the clinically relevant compound pyrimethamine. We identify T. gondii HDAC3 (TgHDAC3) as the target of FR235222 in Toxoplasma tachyzoites and demonstrate the crucial role of the conserved and Apicomplexa HDAC-specific residue TgHDAC3 T99 in the inhibitory activity of the drug. We also show that FR235222 induces differentiation of the tachyzoite (replicative) into the bradyzoite (nonreplicative) stage. Additionally, via its anti-TgHDAC3 activity, FR235222 influences the expression of approximately 370 genes, a third of which are stage-specifically expressed. These results identify FR235222 as a potent HDACi of Apicomplexa, and establish HDAC3 as a central regulator of gene expression and stage conversion in Toxoplasma and, likely, other Apicomplexa.


Assuntos
Apicomplexa/genética , Histona Desacetilases/genética , Animais , Apicomplexa/citologia , Apicomplexa/enzimologia , Diferenciação Celular , Sequência Conservada , Regulação da Expressão Gênica , Inibidores de Histona Desacetilases , Histona Desacetilases/efeitos dos fármacos , Peptídeos Cíclicos/farmacologia , Plasmodium/enzimologia , Plasmodium/genética , Toxoplasma/enzimologia , Toxoplasma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...