Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS Pathog ; 20(6): e1012360, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38935780

RESUMEN

The cGMP-dependent protein kinase (PKG) is the sole cGMP sensor in malaria parasites, acting as an essential signalling hub to govern key developmental processes throughout the parasite life cycle. Despite the importance of PKG in the clinically relevant asexual blood stages, many aspects of malarial PKG regulation, including the importance of phosphorylation, remain poorly understood. Here we use genetic and biochemical approaches to show that reduced cGMP binding to cyclic nucleotide binding domain B does not affect in vitro kinase activity but prevents parasite egress. Similarly, we show that phosphorylation of a key threonine residue (T695) in the activation loop is dispensable for kinase activity in vitro but is essential for in vivo PKG function, with loss of T695 phosphorylation leading to aberrant phosphorylation events across the parasite proteome and changes to the substrate specificity of PKG. Our findings indicate that Plasmodium PKG is uniquely regulated to transduce signals crucial for malaria parasite development.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico , GMP Cíclico , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Fosforilación , GMP Cíclico/metabolismo , Malaria/parasitología , Malaria/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Animales , Plasmodium falciparum/metabolismo , Plasmodium falciparum/genética , Humanos , Transducción de Señal , Eritrocitos/parasitología , Eritrocitos/metabolismo
2.
PLoS Pathog ; 19(6): e1011449, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37352369

RESUMEN

Malaria parasite release (egress) from host red blood cells involves parasite-mediated membrane poration and rupture, thought to involve membrane-lytic effector molecules such as perforin-like proteins and/or phospholipases. With the aim of identifying these effectors, we disrupted the expression of two Plasmodium falciparum perforin-like proteins simultaneously and showed that they have no essential roles during blood stage egress. Proteomic profiling of parasite proteins discharged into the parasitophorous vacuole (PV) just prior to egress detected the presence in the PV of a lecithin:cholesterol acyltransferase (LCAT; PF3D7_0629300). Conditional ablation of LCAT resulted in abnormal egress and a reduced replication rate. Lipidomic profiles of LCAT-null parasites showed drastic changes in several phosphatidylserine and acylphosphatidylglycerol species during egress. We thus show that, in addition to its previously demonstrated role in liver stage merozoite egress, LCAT is required to facilitate efficient egress in asexual blood stage malaria parasites.


Asunto(s)
Malaria Falciparum , Malaria , Parásitos , Animales , Parásitos/metabolismo , Fosfolipasas , Perforina , Proteómica , Eritrocitos/parasitología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Malaria Falciparum/parasitología
3.
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
4.
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
5.
Sci Adv ; 7(13)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33762339

RESUMEN

Calcium signaling regulated by the cGMP-dependent protein kinase (PKG) controls key life cycle transitions in the malaria parasite. However, how calcium is mobilized from intracellular stores in the absence of canonical calcium channels in Plasmodium is unknown. Here, we identify a multipass membrane protein, ICM1, with homology to transporters and calcium channels that is tightly associated with PKG in both asexual blood stages and transmission stages. Phosphoproteomic analyses reveal multiple ICM1 phosphorylation events dependent on PKG activity. Stage-specific depletion of Plasmodium berghei ICM1 prevents gametogenesis due to a block in intracellular calcium mobilization, while conditional loss of Plasmodium falciparum ICM1 is detrimental for the parasite resulting in severely reduced calcium mobilization, defective egress, and lack of invasion. Our findings suggest that ICM1 is a key missing link in transducing PKG-dependent signals and provide previously unknown insights into atypical calcium homeostasis in malaria parasites essential for pathology and disease transmission.


Asunto(s)
Malaria , Parásitos , Animales , Calcio/metabolismo , Canales de Calcio , Gametogénesis , Malaria/parasitología , Proteínas de la Membrana/metabolismo , Plasmodium berghei/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
6.
Life Sci Alliance ; 3(4)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32179592

RESUMEN

Over recent years, a plethora of new genetic tools has transformed conditional engineering of the malaria parasite genome, allowing functional dissection of essential genes in the asexual and sexual blood stages that cause pathology or are required for disease transmission, respectively. Important challenges remain, including the desirability to complement conditional mutants with a correctly regulated second gene copy to confirm that observed phenotypes are due solely to loss of gene function and to analyse structure-function relationships. To meet this challenge, here we combine the dimerisable Cre (DiCre) system with the use of multiple lox sites to simultaneously generate multiple recombination events of the same gene. We focused on the Plasmodium falciparum cGMP-dependent protein kinase (PKG), creating in parallel conditional disruption of the gene plus up to two allelic replacements. We use the approach to demonstrate that PKG has no scaffolding or adaptor role in intraerythrocytic development, acting solely at merozoite egress. We also show that a phosphorylation-deficient PKG is functionally incompetent. Our method provides valuable new tools for analysis of gene function in the malaria parasite.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Plasmodium falciparum/genética , Alelos , Animales , Eritrocitos/metabolismo , Eliminación de Gen , Malaria/genética , Parásitos/metabolismo , Fenotipo , Fosforilación , Proteínas Protozoarias/genética
7.
Biochem J ; 477(2): 525-540, 2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31942933

RESUMEN

Subtilisin-like serine peptidases (subtilases) play important roles in the life cycle of many organisms, including the protozoan parasites that are the causative agent of malaria, Plasmodium spp. As with other peptidases, subtilase proteolytic activity has to be tightly regulated in order to prevent potentially deleterious uncontrolled protein degradation. Maturation of most subtilases requires the presence of an N-terminal propeptide that facilitates folding of the catalytic domain. Following its proteolytic cleavage, the propeptide acts as a transient, tightly bound inhibitor until its eventual complete removal to generate active protease. Here we report the identification of a stand-alone malaria parasite propeptide-like protein, called SUB1-ProM, encoded by a conserved gene that lies in a highly syntenic locus adjacent to three of the four subtilisin-like genes in the Plasmodium genome. Template-based modelling and ab initio structure prediction showed that the SUB1-ProM core structure is most similar to the X-ray crystal structure of the propeptide of SUB1, an essential parasite subtilase that is discharged into the parasitophorous vacuole (PV) to trigger parasite release (egress) from infected host cells. Recombinant Plasmodium falciparum SUB1-ProM was found to be a fast-binding, potent inhibitor of P. falciparum SUB1, but not of the only other essential blood-stage parasite subtilase, SUB2, or of other proteases examined. Mass-spectrometry and immunofluorescence showed that SUB1-ProM is expressed in the PV of blood stage P. falciparum, where it may act as an endogenous inhibitor to regulate SUB1 activity in the parasite.


Asunto(s)
Malaria Falciparum/genética , Plasmodium falciparum/genética , Serina Proteasas/química , Subtilisina/genética , Secuencia de Aminoácidos/genética , Animales , Eritrocitos/parasitología , Genoma/genética , Humanos , Estadios del Ciclo de Vida/genética , Malaria Falciparum/enzimología , Malaria Falciparum/parasitología , Péptido Hidrolasas/química , Péptido Hidrolasas/genética , Plasmodium falciparum/patogenicidad , Proteolisis , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Serina Proteasas/genética , Subtilisina/química , Vacuolas/parasitología
8.
Insect Biochem Mol Biol ; 98: 48-61, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29751047

RESUMEN

In this work we report a fast and efficient virtual screening protocol for discovery of novel bioinspired synthetic mosquito repellents with lower volatility and, in all likelihood, increased protection time as compared with their plant-derived parental compounds. Our screening protocol comprises two filtering steps. The first filter is based on the shape and chemical similarity to known plant-derived repellents, whereas the second filter is based on the predicted similarity of the ligand's binding mode to the Anopheles gambiae odorant binding protein (AgamOBP1) relative to that of DEET and Icaridin to the same OBP. Using this protocol, a chemical library containing 42,755 synthetic molecules was screened in silico and sixteen selected compounds were tested for their affinity to AgamOBP1 in vitro and repellence against A. gambiae female mosquitoes using a warm-body repellent assay. One of them showed DEET-like repellence (91%) but with significantly lower volatility (2.84 × 10-6 mmHg) than either DEET (1.35 × 10-3 mmHg) or its parental cuminic acid (3.08 × 10-3 mmHg), and four other compounds were found to exhibit repellent indices between 69 and 79%. Overall, a correlation was not evident between repellence and OBP-binding strength. In contrast, a correlation between binding mode and repellence was found.


Asunto(s)
Descubrimiento de Drogas/métodos , Repelentes de Insectos/análisis , Receptores Odorantes/agonistas , Animales , Culicidae , Femenino , Cobayas , Ligandos , Simulación del Acoplamiento Molecular , Bibliotecas de Moléculas Pequeñas
9.
Insect Biochem Mol Biol ; 96: 36-50, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29656020

RESUMEN

There is increasing interest in the development of effective mosquito repellents of natural origin to reduce transmission of diseases such as malaria and yellow fever. To achieve this we have employed an in vitro competition assay involving odorant-binding proteins (OBPs) of the malaria mosquito, Anopheles gambiae, with a predominantly female expression bias to identify plant essential oils (EOs) containing bioactive compounds that target mosquito olfactory function. EOs and their fractions capable of binding to such OBPs displayed repellence against female mosquitoes in a laboratory repellent assay. Repellent EOs were subjected to gas chromatographic analysis linked to antennogram (EAG) recordings from female A. gambiae to identify the biologically active constituents. Among these compounds cumin alcohol, carvacrol, ethyl cinnamate and butyl cinnamate proved as effective as DEET at an equivalent dose in the repellent assay, and combinations of carvacrol with either butyl cinnamate or cumin alcohol proved to be significantly more effective than DEET in the assay. When tested as spatial repellents in experimental shelters housing sleeping humans in northern Nigeria a binary mixture of carvacrol plus cumin alcohol caused mosquitoes to leave shelters in significantly higher numbers to those induced by DEET in female Anopheles spp. and in numbers equivalent to that of DEET in Culex spp. mosquitoes. These findings indicate an approach for the identification of biologically active molecules of natural origin serving as repellents for mosquitoes.


Asunto(s)
Anopheles , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas de Insectos , Repelentes de Insectos/farmacología , Receptores Odorantes , Caracteres Sexuales , Animales , Anopheles/genética , Anopheles/metabolismo , Regulación de la Expresión Génica/fisiología , Proteínas de Insectos/biosíntesis , Proteínas de Insectos/genética , Receptores Odorantes/biosíntesis , Receptores Odorantes/genética
10.
Mol Biochem Parasitol ; 220: 10-14, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29305880

RESUMEN

In an effort to eradicate malaria, new interventions are proposed to include compound/vaccine development against pre-erythrocytic, erythrocytic and mosquito stages of Plasmodium. Drug repurposing might be an alternative approach to new antimalarials reducing the cost and the time required for drug development. Previous in vitro studies have examined the effects of protease inhibitors on different stages of the Plasmodium parasite, although the clinical relevance of this remains unclear. In this study we tested the putative effect of three HIV protease inhibitors, two general aspartyl protease inhibitors and three AAA-p97 ATPase inhibitors on the zygote to ookinete transition of the Plasmodium parasite. Apart from the two general aspartyl inhibitors, all other compounds had a profound effect on the development of the parasites. HIVPIs inhibited zygote to ookinete conversion by 75%-90%, while the three AAA-p97 ATPase inhibitors blocked conversion by 50%-90% at similar concentrations, while electron microscopy highlighted nuclear and structural abnormalities. Our results highlight a potential of HIV protease inhibitors and p97 inhibitors as transmission blocking agents for the eradication of malaria.


Asunto(s)
Antimaláricos/farmacología , Reposicionamiento de Medicamentos , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/crecimiento & desarrollo , Inhibidores de Proteasas/farmacología , Pruebas de Sensibilidad Parasitaria
11.
PLoS One ; 12(1): e0170260, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28107409

RESUMEN

Site-2 proteases (S2P) belong to the M50 family of metalloproteases, which typically perform essential roles by mediating activation of membrane-bound transcription factors through regulated intramembrane proteolysis (RIP). Protease-dependent liberation of dormant transcription factors triggers diverse cellular responses, such as sterol regulation, Notch signalling and the unfolded protein response. Plasmodium parasites rely on regulated proteolysis for controlling essential pathways throughout the life cycle. In this study we examine the Plasmodium-encoded S2P in a murine malaria model and show that it is expressed in all stages of Plasmodium development. Localisation studies by endogenous gene tagging revealed that in all invasive stages the protein is in close proximity to the nucleus. Ablation of PbS2P by reverse genetics leads to reduced growth rates during liver and blood infection and, hence, virulence attenuation. Strikingly, absence of PbS2P was compatible with parasite life cycle progression in the mosquito and mammalian hosts under physiological conditions, suggesting redundant or dispensable roles in vivo.


Asunto(s)
Malaria/enzimología , Péptido Hidrolasas/genética , Plasmodium/enzimología , Secuencia de Aminoácidos , Animales , Núcleo Celular/enzimología , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Hígado/parasitología , Ratones , Ratones Endogámicos C57BL , Péptido Hidrolasas/química , Plasmodium/genética , Homología de Secuencia de Aminoácido
12.
PLoS One ; 10(3): e0121379, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25803874

RESUMEN

Regulated proteolysis is known to control a variety of vital processes in apicomplexan parasites including invasion and egress of host cells. Serine proteases have been proposed as targets for drug development based upon inhibitor studies that show parasite attenuation and transmission blockage. Genetic studies suggest that serine proteases, such as subtilisin and rhomboid proteases, are essential but functional studies have proved challenging as active proteases are difficult to express. Proteinaceous Protease Inhibitors (PPIs) provide an alternative way to address the role of serine proteases in apicomplexan biology. To validate such an approach, a Neospora caninum Kazal inhibitor (NcPI-S) was expressed ectopically in two apicomplexan species, Toxoplasma gondii tachyzoites and Plasmodium berghei ookinetes, with the aim to disrupt proteolytic processes taking place within the secretory pathway. NcPI-S negatively affected proliferation of Toxoplasma tachyzoites, while it had no effect on invasion and egress. Expression of the inhibitor in P. berghei zygotes blocked their development into mature and invasive ookinetes. Moreover, ultra-structural studies indicated that expression of NcPI-S interfered with normal formation of micronemes, which was also confirmed by the lack of expression of the micronemal protein SOAP in these parasites. Our results suggest that NcPI-S could be a useful tool to investigate the function of proteases in processes fundamental for parasite survival, contributing to the effort to identify targets for parasite attenuation and transmission blockage.


Asunto(s)
Neospora/metabolismo , Plasmodium berghei/crecimiento & desarrollo , Inhibidores de Serina Proteinasa/metabolismo , Toxoplasma/crecimiento & desarrollo , Clonación Molecular , Cartilla de ADN/genética , Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Expresión Génica Ectópica/fisiología , Técnica del Anticuerpo Fluorescente Indirecta , Immunoblotting , Microscopía Electrónica de Transmisión , Plásmidos/genética , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/metabolismo , Proteolisis , Inhibidores de Serina Proteinasa/farmacología , Toxoplasma/efectos de los fármacos , Toxoplasma/metabolismo , Transfección
13.
J Biol Chem ; 290(12): 7961-72, 2015 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-25657000

RESUMEN

The identification of molecular targets of insect repellents has been a challenging task, with their effects on odorant receptors (ORs) remaining a debatable issue. Here, we describe a study on the effects of selected mosquito repellents, including the widely used repellent N,N-diethyl-meta-toluamide (DEET), on the function of specific ORs of the African malaria vector Anopheles gambiae. This study, which has been based on quantitative measurements of a Ca(2+)-activated photoprotein biosensor of recombinant OR function in an insect cell-based expression platform and a sequential compound addition protocol, revealed that heteromeric OR (ORx/Orco) function was susceptible to strong inhibition by all tested mosquito repellents except DEET. Moreover, our results demonstrated that the observed inhibition was due to efficient blocking of Orco (olfactory receptor coreceptor) function. This mechanism of repellent action, which is reported for the first time, is distinct from the mode of action of other characterized insect repellents including DEET.


Asunto(s)
Anopheles/fisiología , Repelentes de Insectos , Receptores Odorantes/metabolismo , Animales , Anopheles/metabolismo
14.
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
15.
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
16.
Subcell Biochem ; 47: 121-39, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18512347

RESUMEN

Apicomplexan pathogens replicate exclusively within the confines of a host cell. Entry into (invasion) and exit from (egress) these cells requires an array of specialized parasite molecules, many of which have long been considered to have potential as targets of drug or vaccine-based therapies. In this chapter the authors discuss the current state of knowledge regarding the role of parasite proteolytic enzymes in these critical steps in the life cycle of two clinically important apicomplexan genera, Plasmodium and Toxoplasma. At least three distinct proteases of the cysteine mechanistic class have been implicated in egress of the malaria parasite from cells of its vertebrate and insect host. In contrast, the bulk of the evidence indicates a prime role for serine proteases of the subtilisin and rhomboid families in invasion by both parasites. Whereas proteases involved in egress may function predominantly to degrade host cell structures, proteases involved in invasion probably act primarily as maturases and 'sheddases', required to activate and ultimately remove ligands involved in interactions with the host cell.


Asunto(s)
Péptido Hidrolasas/metabolismo , Plasmodium/enzimología , Toxoplasma/enzimología , Animales , Interacciones Huésped-Parásitos , Humanos , Modelos Biológicos , Péptido Hidrolasas/química , Péptido Hidrolasas/clasificación , Plasmodium/genética , Plasmodium/metabolismo , Plasmodium/patogenicidad , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasma/patogenicidad
17.
Cell ; 131(6): 1072-83, 2007 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-18083098

RESUMEN

The most virulent form of malaria is caused by waves of replication of blood stages of the protozoan pathogen Plasmodium falciparum. The parasite divides within an intraerythrocytic parasitophorous vacuole until rupture of the vacuole and host-cell membranes releases merozoites that invade fresh erythrocytes to repeat the cycle. Despite the importance of merozoite egress for disease progression, none of the molecular factors involved are known. We report that, just prior to egress, an essential serine protease called PfSUB1 is discharged from previously unrecognized parasite organelles (termed exonemes) into the parasitophorous vacuole space. There, PfSUB1 mediates the proteolytic maturation of at least two essential members of another enzyme family called SERA. Pharmacological blockade of PfSUB1 inhibits egress and ablates the invasive capacity of released merozoites. Our findings reveal the presence in the malarial parasitophorous vacuole of a regulated, PfSUB1-mediated proteolytic processing event required for release of viable parasites from the host erythrocyte.


Asunto(s)
Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Malaria/parasitología , Plasmodium falciparum/enzimología , Proteínas Protozoarias/fisiología , Subtilisinas/fisiología , Animales , Antígenos de Protozoos/metabolismo , Antígenos de Protozoos/fisiología , Estadios del Ciclo de Vida , Malaria/sangre , Modelos Biológicos , Plasmodium falciparum/patogenicidad , Plasmodium falciparum/ultraestructura , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/aislamiento & purificación , Proteínas Protozoarias/metabolismo , Esporozoítos/fisiología , Subtilisinas/antagonistas & inhibidores , Subtilisinas/aislamiento & purificación , Subtilisinas/metabolismo , Vacuolas/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA