Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
EMBO J ; 40(11): e107226, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33932049

RESUMEN

Malaria parasite egress from host erythrocytes (RBCs) is regulated by discharge of a parasite serine protease called SUB1 into the parasitophorous vacuole (PV). There, SUB1 activates a PV-resident cysteine protease called SERA6, enabling host RBC rupture through SERA6-mediated degradation of the RBC cytoskeleton protein ß-spectrin. Here, we show that the activation of Plasmodium falciparum SERA6 involves a second, autocatalytic step that is triggered by SUB1 cleavage. Unexpectedly, autoproteolytic maturation of SERA6 requires interaction in multimolecular complexes with a distinct PV-located protein cofactor, MSA180, that is itself a SUB1 substrate. Genetic ablation of MSA180 mimics SERA6 disruption, producing a fatal block in ß-spectrin cleavage and RBC rupture. Drug-like inhibitors of SERA6 autoprocessing similarly prevent ß-spectrin cleavage and egress in both P. falciparum and the emerging zoonotic pathogen P. knowlesi. Our results elucidate the egress pathway and identify SERA6 as a target for a new class of antimalarial drugs designed to prevent disease progression.


Asunto(s)
Antimaláricos/farmacología , Proteasas de Cisteína/metabolismo , Plasmodium falciparum/metabolismo , Inhibidores de Proteasas/farmacología , Proteínas Protozoarias/metabolismo , Células Cultivadas , Eritrocitos/metabolismo , Eritrocitos/parasitología , Humanos , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/patogenicidad , Proteolisis , Proteínas Protozoarias/antagonistas & inhibidores , Serina Proteasas/metabolismo , Espectrina/metabolismo
2.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33975947

RESUMEN

Malaria is a devastating infectious disease, which causes over 400,000 deaths per annum and impacts the lives of nearly half the world's population. The causative agent, a protozoan parasite, replicates within red blood cells (RBCs), eventually destroying the cells in a lytic process called egress to release a new generation of parasites. These invade fresh RBCs to repeat the cycle. Egress is regulated by an essential parasite subtilisin-like serine protease called SUB1. Here, we describe the development and optimization of substrate-based peptidic boronic acids that inhibit Plasmodium falciparum SUB1 with low nanomolar potency. Structural optimization generated membrane-permeable, slow off-rate inhibitors that prevent Pfalciparum egress through direct inhibition of SUB1 activity and block parasite replication in vitro at submicromolar concentrations. Our results validate SUB1 as a potential target for a new class of antimalarial drugs designed to prevent parasite replication and disease progression.


Asunto(s)
Antimaláricos/farmacología , Ácidos Borónicos/farmacología , Péptidos/farmacología , Plasmodium falciparum/efectos de los fármacos , Proteínas Protozoarias/química , Subtilisinas/química , Antimaláricos/síntesis química , Sitios de Unión , Ácidos Borónicos/síntesis química , Diseño de Fármacos , Eritrocitos/efectos de los fármacos , Eritrocitos/parasitología , Expresión Génica , Humanos , Cinética , Estadios del Ciclo de Vida/efectos de los fármacos , Estadios del Ciclo de Vida/fisiología , Modelos Moleculares , Simulación del Acoplamiento Molecular , Péptidos/síntesis química , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Relación Estructura-Actividad , Especificidad por Sustrato , Subtilisinas/antagonistas & inhibidores , Subtilisinas/genética , Subtilisinas/metabolismo , Termodinámica
3.
Proc Natl Acad Sci U S A ; 114(13): 3439-3444, 2017 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-28292906

RESUMEN

In the asexual blood stages of malarial infection, merozoites invade erythrocytes and replicate within a parasitophorous vacuole to form daughter cells that eventually exit (egress) by sequential rupture of the vacuole and erythrocyte membranes. The current model is that PKG, a malarial cGMP-dependent protein kinase, triggers egress, activating malarial proteases and other effectors. Using selective inhibitors of either PKG or cysteine proteases to separately inhibit the sequential steps in membrane perforation, combined with video microscopy, electron tomography, electron energy loss spectroscopy, and soft X-ray tomography of mature intracellular Plasmodium falciparum parasites, we resolve intermediate steps in egress. We show that the parasitophorous vacuole membrane (PVM) is permeabilized 10-30 min before its PKG-triggered breakdown into multilayered vesicles. Just before PVM breakdown, the host red cell undergoes an abrupt, dramatic shape change due to the sudden breakdown of the erythrocyte cytoskeleton, before permeabilization and eventual rupture of the erythrocyte membrane to release the parasites. In contrast to the previous view of PKG-triggered initiation of egress and a gradual dismantling of the host erythrocyte cytoskeleton over the course of schizont development, our findings identify an initial step in egress and show that host cell cytoskeleton breakdown is restricted to a narrow time window within the final stages of egress.


Asunto(s)
Citoesqueleto/metabolismo , Membrana Eritrocítica/parasitología , Eritrocitos/parasitología , Malaria Falciparum/parasitología , Plasmodium falciparum/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Citoesqueleto/genética , Membrana Eritrocítica/metabolismo , Eritrocitos/metabolismo , Humanos , Malaria Falciparum/genética , Malaria Falciparum/metabolismo , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
4.
PLoS Pathog ; 13(7): e1006453, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28683142

RESUMEN

Egress of the malaria parasite Plasmodium falciparum from its host red blood cell is a rapid, highly regulated event that is essential for maintenance and completion of the parasite life cycle. Egress is protease-dependent and is temporally associated with extensive proteolytic modification of parasite proteins, including a family of papain-like proteins called SERA that are expressed in the parasite parasitophorous vacuole. Previous work has shown that the most abundant SERA, SERA5, plays an important but non-enzymatic role in asexual blood stages. SERA5 is extensively proteolytically processed by a parasite serine protease called SUB1 as well as an unidentified cysteine protease just prior to egress. However, neither the function of SERA5 nor the role of its processing is known. Here we show that conditional disruption of the SERA5 gene, or of both the SERA5 and related SERA4 genes simultaneously, results in a dramatic egress and replication defect characterised by premature host cell rupture and the failure of daughter merozoites to efficiently disseminate, instead being transiently retained within residual bounding membranes. SERA5 is not required for poration (permeabilization) or vesiculation of the host cell membrane at egress, but the premature rupture phenotype requires the activity of a parasite or host cell cysteine protease. Complementation of SERA5 null parasites by ectopic expression of wild-type SERA5 reversed the egress defect, whereas expression of a SERA5 mutant refractory to processing failed to rescue the phenotype. Our findings implicate SERA5 as an important regulator of the kinetics and efficiency of egress and suggest that proteolytic modification is required for SERA5 function. In addition, our study reveals that efficient egress requires tight control of the timing of membrane rupture.


Asunto(s)
Antígenos de Protozoos/metabolismo , Eritrocitos/parasitología , Malaria Falciparum/parasitología , Péptido Hidrolasas/metabolismo , Plasmodium falciparum/fisiología , Animales , Antígenos de Protozoos/genética , Membrana Celular/parasitología , Eritrocitos/química , Humanos , Cinética , Merozoítos/química , Merozoítos/genética , Merozoítos/crecimiento & desarrollo , Merozoítos/fisiología , Péptido Hidrolasas/genética , Plasmodium falciparum/química , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Proteolisis
5.
Blood ; 127(3): 343-51, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26637786

RESUMEN

Much of the virulence of Plasmodium falciparum malaria is caused by cytoadherence of infected erythrocytes, which promotes parasite survival by preventing clearance in the spleen. Adherence is mediated by membrane protrusions known as knobs, whose formation depends on the parasite-derived, knob-associated histidine-rich protein (KAHRP). Knobs are required for cytoadherence under flow conditions, and they contain both KAHRP and the parasite-derived erythrocyte membrane protein PfEMP1. Using electron tomography, we have examined the 3-dimensional structure of knobs in detergent-insoluble skeletons of P falciparum 3D7 schizonts. We describe a highly organized knob skeleton composed of a spiral structure coated by an electron-dense layer underlying the knob membrane. This knob skeleton is connected by multiple links to the erythrocyte cytoskeleton. We used immuno-electron microscopy (EM) to locate KAHRP in these structures. The arrangement of membrane proteins in the knobs, visualized by high-resolution freeze-fracture scanning EM, is distinct from that in the surrounding erythrocyte membrane, with a structure at the apex that likely represents the adhesion site. Thus, erythrocyte knobs in P falciparum infection contain a highly organized skeleton structure underlying a specialized region of membrane. We propose that the spiral and dense coat organize the cytoadherence structures in the knob, and anchor them into the erythrocyte cytoskeleton. The high density of knobs and their extensive mechanical linkage suggest an explanation for the rigidification of the cytoskeleton in infected cells, and for the transmission to the cytoskeleton of shear forces experienced by adhering cells.


Asunto(s)
Eritrocitos/parasitología , Eritrocitos/ultraestructura , Malaria Falciparum/patología , Malaria Falciparum/parasitología , Plasmodium falciparum/fisiología , Citoesqueleto/metabolismo , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/ultraestructura , Eritrocitos/metabolismo , Humanos , Proteínas de la Membrana/metabolismo
6.
Mol Microbiol ; 96(2): 368-87, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25599609

RESUMEN

The malaria parasite Plasmodium falciparum replicates in an intraerythrocytic parasitophorous vacuole (PV). The most abundant P. falciparum PV protein, called SERA5, is essential in blood stages and possesses a papain-like domain, prompting speculation that it functions as a proteolytic enzyme. Unusually however, SERA5 possesses a Ser residue (Ser596) at the position of the canonical catalytic Cys of papain-like proteases, and the function of SERA5 or whether it performs an enzymatic role is unknown. In this study, we failed to detect proteolytic activity associated with the Ser596-containing parasite-derived or recombinant protein. However, substitution of Ser596 with a Cys residue produced an active recombinant enzyme with characteristics of a cysteine protease, demonstrating that SERA5 can bind peptides. Using targeted homologous recombination in P. falciparum, we substituted Ser596 with Ala with no phenotypic consequences, proving that SERA5 does not perform an essential enzymatic role in the parasite. We could also replace an internal segment of SERA5 with an affinity-purification tag. In contrast, using almost identical targeting constructs, we could not truncate or C-terminally tag the SERA5 gene, or replace Ser596 with a bulky Arg residue. Our findings show that SERA5 plays an indispensable but non-enzymatic role in the P. falciparum blood-stage life cycle.


Asunto(s)
Antígenos de Protozoos/metabolismo , Malaria Falciparum/parasitología , Péptido Hidrolasas/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Secuencias de Aminoácidos , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Humanos , Estadios del Ciclo de Vida , Malaria Falciparum/sangre , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Plasmodium falciparum/fisiología , Reproducción Asexuada
7.
PLoS Pathog ; 9(5): e1003344, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23675297

RESUMEN

The malaria parasite replicates within an intraerythrocytic parasitophorous vacuole (PV). Eventually, in a tightly regulated process called egress, proteins of the PV and intracellular merozoite surface are modified by an essential parasite serine protease called PfSUB1, whilst the enclosing PV and erythrocyte membranes rupture, releasing merozoites to invade fresh erythrocytes. Inhibition of the Plasmodium falciparum cGMP-dependent protein kinase (PfPKG) prevents egress, but the underlying mechanism is unknown. Here we show that PfPKG activity is required for PfSUB1 discharge into the PV, as well as for release of distinct merozoite organelles called micronemes. Stimulation of PfPKG by inhibiting parasite phosphodiesterase activity induces premature PfSUB1 discharge and egress of developmentally immature, non-invasive parasites. Our findings identify the signalling pathway that regulates PfSUB1 function and egress, and raise the possibility of targeting PfPKG or parasite phosphodiesterases in therapeutic approaches to dysregulate critical protease-mediated steps in the parasite life cycle.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Interacciones Huésped-Parásitos/fisiología , Merozoítos/fisiología , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Transducción de Señal/fisiología , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Orgánulos/metabolismo
8.
Mol Microbiol ; 88(4): 687-701, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23489321

RESUMEN

Asexual blood stages of the malaria parasite, which cause all the pathology associated with malaria, can readily be genetically modified by homologous recombination, enabling the functional study of parasite genes that are not essential in this part of the life cycle. However, no widely applicable method for conditional mutagenesis of essential asexual blood-stage malarial genes is available, hindering their functional analysis. We report the application of the DiCre conditional recombinase system to Plasmodium falciparum, the causative agent of the most dangerous form of malaria. We show that DiCre can be used to obtain rapid, highly regulated site-specific recombination in P. falciparum, capable of excising loxP-flanked sequences from a genomic locus with close to 100% efficiency within the time-span of a single erythrocytic growth cycle. DiCre-mediated deletion of the SERA5 3' UTR failed to reduce expression of the gene due to the existence of alternative cryptic polyadenylation sites within the modified locus. However, we successfully used the system to recycle the most widely used drug resistance marker for P. falciparum, human dihydrofolate reductase, in the process producing constitutively DiCre-expressing P. falciparum clones that have broad utility for the functional analysis of essential asexual blood-stage parasite genes.


Asunto(s)
Eliminación de Gen , Genética Microbiana/métodos , Integrasas/metabolismo , Biología Molecular/métodos , Parasitología/métodos , Plasmodium falciparum/genética , Expresión Génica , Genes Protozoarios , Integrasas/genética , Plasmodium falciparum/crecimiento & desarrollo , Recombinación Genética
9.
Biochim Biophys Acta Gen Subj ; 1868(9): 130665, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38969256

RESUMEN

BACKGROUND: The malaria parasite Plasmodium falciparum replicates within red blood cells, then ruptures the cell in a process called egress in order to continue its life cycle. Egress is regulated by a proteolytic cascade involving an essential parasite subtilisin-like serine protease called SUB1. Maturation of SUB1 initiates in the parasite endoplasmic reticulum with autocatalytic cleavage of an N-terminal prodomain (p31), which initially remains non-covalently bound to the catalytic domain, p54. Further trafficking of the p31-p54 complex results in formation of a terminal p47 form of the SUB1 catalytic domain. Recent work has implicated a parasite aspartic protease, plasmepsin X (PMX), in maturation of the SUB1 p31-p54 complex through controlled cleavage of the prodomain p31. METHODS: Here we use biochemical and enzymatic analysis to examine the activation of SUB1 by PMX. RESULTS: We show that both p31 and p31-p54 are largely dimeric under the relatively acidic conditions to which they are likely exposed to PMX in the parasite. We confirm the sites within p31 that are cleaved by PMX and determine the order of cleavage. We find that cleavage by PMX results in rapid loss of the capacity of p31 to act as an inhibitor of SUB1 catalytic activity and we directly demonstrate that exposure to PMX of recombinant p31-p54 complex activates SUB1 activity. CONCLUSIONS: Our results confirm that precise, PMX-mediated cleavage of the SUB1 prodomain activates SUB1 enzyme activity. GENERAL SIGNIFICANCE: Our findings elucidate the role of PMX in activation of SUB1, a key effector of malaria parasite egress.

10.
bioRxiv ; 2024 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-38260474

RESUMEN

Malaria, caused by Plasmodium falciparum, remains a significant health burden. A barrier for developing anti-malarial drugs is the ability of the parasite to rapidly generate resistance. We demonstrated that Salinipostin A (SalA), a natural product, kills parasites by inhibiting multiple lipid metabolizing serine hydrolases, a mechanism with a low propensity for resistance. Given the difficulty of employing natural products as therapeutic agents, we synthesized a library of lipidic mixed alkyl/aryl phosphonates as bioisosteres of SalA. Two constitutional isomers exhibited divergent anti-parasitic potencies which enabled identification of therapeutically relevant targets. We also confirm that this compound kills parasites through a mechanism that is distinct from both SalA and the pan-lipase inhibitor, Orlistat. Like SalA, our compound induces only weak resistance, attributable to mutations in a single protein involved in multidrug resistance. These data suggest that mixed alkyl/aryl phosphonates are a promising, synthetically tractable anti-malarials with a low-propensity to induce resistance.

11.
J Biol Chem ; 287(45): 37949-63, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22984267

RESUMEN

The malaria parasite replicates within an intraerythrocytic parasitophorous vacuole (PV). The PV and host cell membranes eventually rupture, releasing merozoites in a process called egress. Certain inhibitors of serine and cysteine proteases block egress, indicating a crucial role for proteases. The Plasmodium falciparum genome encodes nine serine-repeat antigens (SERAs), each of which contains a central domain homologous to the papain-like (clan CA, family C1) protease family. SERA5 and SERA6 are indispensable in blood-stage parasites, but the function of neither is known. Here we show that SERA6 localizes to the PV where it is precisely cleaved just prior to egress by an essential serine protease called PfSUB1. Mutations that replace the predicted catalytic Cys of SERA6, or that block SERA6 processing by PfSUB1, could not be stably introduced into the parasite genomic sera6 locus, indicating that SERA6 is an essential enzyme and that processing is important for its function. We demonstrate that cleavage of SERA6 by PfSUB1 converts it to an active cysteine protease. Our observations reveal a proteolytic activation step in the malarial PV that may be required for release of the parasite from its host erythrocyte.


Asunto(s)
Proteasas de Cisteína/metabolismo , Plasmodium falciparum/enzimología , Proteínas Protozoarias/metabolismo , Subtilisinas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión/genética , Western Blotting , Proteasas de Cisteína/genética , Activación Enzimática , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Microscopía Inmunoelectrónica , Datos de Secuencia Molecular , Mutación , Plasmodium falciparum/genética , Plasmodium falciparum/fisiología , Proteolisis , Proteínas Protozoarias/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Subtilisinas/genética , Vacuolas/enzimología , Vacuolas/ultraestructura
12.
EMBO J ; 28(6): 725-35, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19214190

RESUMEN

The malaria parasite Plasmodium falciparum replicates within an intraerythrocytic parasitophorous vacuole (PV). Rupture of the host cell allows release (egress) of daughter merozoites, which invade fresh erythrocytes. We previously showed that a subtilisin-like protease called PfSUB1 regulates egress by being discharged into the PV in the final stages of merozoite development to proteolytically modify the SERA family of papain-like proteins. Here, we report that PfSUB1 has a further role in 'priming' the merozoite prior to invasion. The major protein complex on the merozoite surface comprises three proteins called merozoite surface protein 1 (MSP1), MSP6 and MSP7. We show that just before egress, all undergo proteolytic maturation by PfSUB1. Inhibition of PfSUB1 activity results in the accumulation of unprocessed MSPs on the merozoite surface, and erythrocyte invasion is significantly reduced. We propose that PfSUB1 is a multifunctional processing protease with an essential role in both egress of the malaria merozoite and remodelling of its surface in preparation for erythrocyte invasion.


Asunto(s)
Eritrocitos/parasitología , Malaria Falciparum/enzimología , Malaria Falciparum/parasitología , Parásitos/enzimología , Plasmodium falciparum/enzimología , Proteínas Protozoarias/metabolismo , Serina Endopeptidasas/metabolismo , Subtilisinas/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Consenso , Eritrocitos/efectos de los fármacos , Humanos , Merozoítos/enzimología , Datos de Secuencia Molecular , Parásitos/efectos de los fármacos , Péptidos/metabolismo , Plasmodium falciparum/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/química , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/química , Inhibidores de Serina Proteinasa/farmacología , Especificidad por Sustrato/efectos de los fármacos , Subtilisinas/antagonistas & inhibidores , Subtilisinas/química
13.
PLoS Pathog ; 7(12): e1002448, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22194692

RESUMEN

The malarial life cycle involves repeated rounds of intraerythrocytic replication interspersed by host cell rupture which releases merozoites that rapidly invade fresh erythrocytes. Apical membrane antigen-1 (AMA1) is a merozoite protein that plays a critical role in invasion. Antibodies against AMA1 prevent invasion and can protect against malaria in vivo, so AMA1 is of interest as a malaria vaccine candidate. AMA1 is efficiently shed from the invading parasite surface, predominantly through juxtamembrane cleavage by a membrane-bound protease called SUB2, but also by limited intramembrane cleavage. We have investigated the structural requirements for shedding of Plasmodium falciparum AMA1 (PfAMA1), and the consequences of its inhibition. Mutagenesis of the intramembrane cleavage site by targeted homologous recombination abolished intramembrane cleavage with no effect on parasite viability in vitro. Examination of PfSUB2-mediated shedding of episomally-expressed PfAMA1 revealed that the position of cleavage is determined primarily by its distance from the parasite membrane. Certain mutations at the PfSUB2 cleavage site block shedding, and parasites expressing these non-cleavable forms of PfAMA1 on a background of expression of the wild type gene invade and replicate normally in vitro. The non-cleavable PfAMA1 is also functional in invasion. However - in contrast to the intramembrane cleavage site - mutations that block PfSUB2-mediated shedding could not be stably introduced into the genomic pfama1 locus, indicating that some shedding of PfAMA1 by PfSUB2 is essential. Remarkably, parasites expressing shedding-resistant forms of PfAMA1 exhibit enhanced sensitivity to antibody-mediated inhibition of invasion. Drugs that inhibit PfSUB2 activity should block parasite replication and may also enhance the efficacy of vaccines based on AMA1 and other merozoite surface proteins.


Asunto(s)
Anticuerpos Antiprotozoarios/farmacología , Antígenos de Protozoos/inmunología , Proteínas de la Membrana/inmunología , Plasmodium falciparum/inmunología , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/farmacología , Antígenos de Protozoos/química , Antígenos de Protozoos/metabolismo , Recombinación Homóloga , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Mutación/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo
14.
J Med Chem ; 66(15): 10658-10680, 2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37505188

RESUMEN

The Plasmodium falciparum aspartic protease plasmepsin X (PMX) is essential for the egress of invasive merozoite forms of the parasite. PMX has therefore emerged as a new potential antimalarial target. Building on peptidic amino alcohols originating from a phenotypic screening hit, we have here developed a series of macrocyclic analogues as PMX inhibitors. Incorporation of an extended linker between the S1 phenyl group and S3 amide led to a lead compound that displayed a 10-fold improved PMX inhibitory potency and a 3-fold improved half-life in microsomal stability assays compared to the acyclic analogue. The lead compound was also the most potent of the new macrocyclic compounds in in vitro parasite growth inhibition. Inhibitor 7k cleared blood-stage P. falciparum in a dose-dependent manner when administered orally to infected humanized mice. Consequently, lead compound 7k represents a promising orally bioavailable molecule for further development as a PMX-targeting antimalarial drug.


Asunto(s)
Antimaláricos , Peptidomiméticos , Ratones , Animales , Antimaláricos/farmacología , Antimaláricos/metabolismo , Peptidomiméticos/farmacología , Peptidomiméticos/metabolismo , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasas/metabolismo , Ácido Aspártico Endopeptidasas , Plasmodium falciparum/metabolismo , Proteínas Protozoarias
15.
J Med Chem ; 65(19): 12535-12545, 2022 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-36137276

RESUMEN

Widespread resistance to many antimalarial therapies currently in use stresses the need for the discovery of new classes of drugs with new modes of action. The subtilisin-like serine protease SUB1 controls egress of malaria parasites (merozoites) from the parasite-infected red blood cell. As such, SUB1 is considered a prospective target for drugs designed to interrupt the asexual blood stage life cycle of the malaria parasite. Inhibitors of SUB1 have potential as wide-spectrum antimalarial drugs, as a single orthologue of SUB1 is found in the genomes of all known Plasmodium species. This mini-perspective provides a short overview of the function and structure of SUB1 and summarizes all of the published SUB1 inhibitors. The inhibitors are classified by the methods of their discovery, including both rational design and screening.


Asunto(s)
Antimaláricos , Malaria , Plasmodium , Antimaláricos/química , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Eritrocitos/metabolismo , Humanos , Malaria/tratamiento farmacológico , Malaria/parasitología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Serina , Inhibidores de Serina Proteinasa , Subtilisinas/química , Subtilisinas/metabolismo
16.
Elife ; 112022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36576255

RESUMEN

The malaria parasite Plasmodium falciparum synthesizes significant amounts of phospholipids to meet the demands of replication within red blood cells. De novo phosphatidylcholine (PC) biosynthesis via the Kennedy pathway is essential, requiring choline that is primarily sourced from host serum lysophosphatidylcholine (lysoPC). LysoPC also acts as an environmental sensor to regulate parasite sexual differentiation. Despite these critical roles for host lysoPC, the enzyme(s) involved in its breakdown to free choline for PC synthesis are unknown. Here, we show that a parasite glycerophosphodiesterase (PfGDPD) is indispensable for blood stage parasite proliferation. Exogenous choline rescues growth of PfGDPD-null parasites, directly linking PfGDPD function to choline incorporation. Genetic ablation of PfGDPD reduces choline uptake from lysoPC, resulting in depletion of several PC species in the parasite, whilst purified PfGDPD releases choline from glycerophosphocholine in vitro. Our results identify PfGDPD as a choline-releasing glycerophosphodiesterase that mediates a critical step in PC biosynthesis and parasite survival.


Malaria kills over half a million people every year worldwide. A single-celled parasite called Plasmodium falciparum is responsible for the most lethal form of the disease. This malaria-causing agent is carried by mosquitos which transmit the parasite to humans through their bite. Once in the bloodstream, the parasite enters red blood cells and starts to replicate so it can go on to infect other cells. Like our cells, P. falciparum is surrounded by a membrane, and further membranes surround a number of its internal compartments. To make these protective coats, the parasite has to gather a nutrient called choline to form an important building block in the membrane. The parasite gets most of its choline by absorbing and digesting a molecule known as lysoPC found in the bloodstream of its host. However, it was unclear precisely how the parasite achieves this. To address this question, Ramaprasad, Burda et al. used genetic and metabolomic approaches to study how P. falciparum breaks down lysoPC. The experiments found that mutant parasites that are unable to make an enzyme called GDPD were able to infect red blood cells, but failed to grow properly once inside the cells. The mutant parasites took up less choline and, as a result, also made fewer membrane building blocks. The team were able to rescue the mutant parasites by supplying them with large quantities of choline, which allowed them to resume growing. Taken together, the findings of Ramaprasad, Burda et al. suggest that P. falciparum uses GDPD to extract choline from lysoPC when it is living in red blood cells. More and more P. falciparum parasites are becoming resistant to many of the drugs currently being used to treat malaria. One solution is to develop new therapies that target different molecules in the parasite. Since it performs such a vital role, GDPD may have the potential to be a future drug target.


Asunto(s)
Malaria Falciparum , Malaria , Parásitos , Animales , Parásitos/metabolismo , Colina/metabolismo , Plasmodium falciparum/genética , Glicerilfosforilcolina/metabolismo , Eritrocitos/parasitología , Malaria Falciparum/parasitología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
17.
Infect Immun ; 79(3): 1086-97, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21220481

RESUMEN

The protozoan pathogen responsible for the most severe form of human malaria, Plasmodium falciparum, replicates asexually in erythrocytes within a membrane-bound parasitophorous vacuole (PV). Following each round of intracellular growth, the PV membrane (PVM) and host cell membrane rupture to release infectious merozoites in a protease-dependent process called egress. Previous work has shown that, just prior to egress, an essential, subtilisin-like parasite protease called PfSUB1 is discharged into the PV lumen, where it directly cleaves a number of important merozoite surface and PV proteins. These include the essential merozoite surface protein complex MSP1/6/7 and members of a family of papain-like putative proteases called SERA (serine-rich antigen) that are implicated in egress. To determine whether PfSUB1 has additional, previously unrecognized substrates, we have performed a bioinformatic and proteomic analysis of the entire late asexual blood stage proteome of the parasite. Our results demonstrate that PfSUB1 is responsible for the proteolytic processing of a range of merozoite, PV, and PVM proteins, including the rhoptry protein RAP1 (rhoptry-associated protein 1) and the merozoite surface protein MSRP2 (MSP7-related protein-2). Our findings imply multiple roles for PfSUB1 in the parasite life cycle, further supporting the case for considering the protease as a potential new antimalarial drug target.


Asunto(s)
Péptido Hidrolasas/metabolismo , Plasmodium falciparum/enzimología , Secuencia de Aminoácidos , Animales , Western Blotting , Electroforesis en Gel de Poliacrilamida , Estadios del Ciclo de Vida/fisiología , Datos de Secuencia Molecular , Péptido Hidrolasas/química , Plasmodium falciparum/crecimiento & desarrollo , Estructura Terciaria de Proteína
18.
PLoS Pathog ; 5(1): e1000273, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19165323

RESUMEN

Host cell invasion by apicomplexan pathogens such as the malaria parasite Plasmodium spp. and Toxoplasma gondii involves discharge of proteins from secretory organelles called micronemes and rhoptries. In Toxoplasma a protein complex comprising the microneme apical membrane antigen 1 (AMA1), two rhoptry neck proteins, and a protein called Ts4705, localises to the moving junction, a region of close apposition between parasite and host cell during invasion. Antibodies against AMA1 prevent invasion and are protective in vivo, and so AMA1 is of widespread interest as a malaria vaccine candidate. Here we report that the AMA1 complex identified in Toxoplasma is conserved in Plasmodium falciparum. We demonstrate that the invasion-inhibitory monoclonal antibody (mAb) 4G2, which recognises P. falciparum AMA1 (PfAMA1), cannot bind when PfAMA1 is in a complex with its partner proteins. We further show that a single completely conserved PfAMA1 residue, Tyr251, lying within a conserved hydrophobic groove adjacent to the mAb 4G2 epitope, is required for complex formation. We propose that mAb 4G2 inhibits invasion by preventing PfAMA1 from interacting with other components of the invasion complex. Our findings should aid the rational design of subunit malaria vaccines based on PfAMA1.


Asunto(s)
Antígenos de Protozoos/inmunología , Plasmodium falciparum/patogenicidad , Toxoplasma/patogenicidad , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/fisiología , Malaria/inmunología , Vacunas contra la Malaria/inmunología , Plasmodium falciparum/inmunología , Toxoplasma/inmunología , Tirosina/fisiología
19.
J Cell Biol ; 174(7): 1023-33, 2006 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-17000879

RESUMEN

Apicomplexan pathogens are obligate intracellular parasites. To enter cells, they must bind with high affinity to host cell receptors and then uncouple these interactions to complete invasion. Merozoites of Plasmodium falciparum, the parasite responsible for the most dangerous form of malaria, invade erythrocytes using a family of adhesins called Duffy binding ligand-erythrocyte binding proteins (DBL-EBPs). The best-characterized P. falciparum DBL-EBP is erythrocyte binding antigen 175 (EBA-175), which binds erythrocyte surface glycophorin A. We report that EBA-175 is shed from the merozoite at around the point of invasion. Shedding occurs by proteolytic cleavage within the transmembrane domain (TMD) at a site that is conserved across the DBL-EBP family. We show that EBA-175 is cleaved by PfROM4, a rhomboid protease that localizes to the merozoite plasma membrane, but not by other rhomboids tested. Mutations within the EBA-175 TMD that abolish cleavage by PfROM4 prevent parasite growth. Our results identify a crucial role for intramembrane proteolysis in the life cycle of this pathogen.


Asunto(s)
Antígenos de Protozoos/fisiología , Membrana Celular/metabolismo , Membrana Eritrocítica/parasitología , Eritrocitos/parasitología , Malaria/parasitología , Plasmodium falciparum/fisiología , Proteínas Protozoarias/fisiología , Animales , Antígenos de Protozoos/genética , Eritrocitos/metabolismo , Interacciones Huésped-Parásitos/fisiología , Humanos , Ligandos , Mutación , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/fisiología
20.
Malar J ; 9: 77, 2010 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-20298576

RESUMEN

BACKGROUND: Plasmodium falciparum merozoites expose at their surface a large protein complex, which is composed of fragments of merozoite surface protein 1 (MSP-1; called MSP-183, MSP-130, MSP-138, and MSP-142) plus associated processing products of MSP-6 and MSP-7. During erythrocyte invasion this complex, as well as an integral membrane protein called apical membrane antigen-1 (AMA-1), is shed from the parasite surface following specific proteolysis. Components of the MSP-1/6/7 complex and AMA-1 are presently under development as malaria vaccines. METHODS: The specificities and effects of antibodies directed against MSP-1, MSP-6, MSP-7 on the growth of blood stage parasites were studied using ELISA and the pLDH-assay. To understand the mode of action of these antibodies, their effects on processing of MSP-1 and AMA-1 on the surface of merozoites were investigated. RESULTS: Antibodies targeting epitopes located throughout the MSP-1/6/7 complex interfere with shedding of MSP-1, and as a consequence prevent erythrocyte invasion. Antibodies targeting the MSP-1/6/7 complex have no effect on the processing and shedding of AMA-1 and, similarly, antibodies blocking the shedding of AMA-1 do not affect cleavage of MSP-1, suggesting completely independent functions of these proteins during invasion. Furthermore, some epitopes, although eliciting highly inhibitory antibodies, are only poorly recognized by the immune system when presented in the structural context of the intact antigen. CONCLUSIONS: The findings reported provide further support for the development of vaccines based on MSP-1/6/7 and AMA-1, which would possibly include a combination of these antigens.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Protozoos/inmunología , Proteínas de la Membrana/inmunología , Proteína 1 de Superficie de Merozoito/inmunología , Plasmodium falciparum/parasitología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Especificidad de Anticuerpos , Antígenos de Superficie/inmunología , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Eritrocitos/metabolismo , Eritrocitos/parasitología , Humanos , Inmunoglobulina G , Vacunas contra la Malaria/inmunología , Ratones , Conejos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA