Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 167(3): 610-624, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27768886

RESUMEN

Malaria has been a major global health problem of humans through history and is a leading cause of death and disease across many tropical and subtropical countries. Over the last fifteen years renewed efforts at control have reduced the prevalence of malaria by over half, raising the prospect that elimination and perhaps eradication may be a long-term possibility. Achievement of this goal requires the development of new tools including novel antimalarial drugs and more efficacious vaccines as well as an increased understanding of the disease and biology of the parasite. This has catalyzed a major effort resulting in development and regulatory approval of the first vaccine against malaria (RTS,S/AS01) as well as identification of novel drug targets and antimalarial compounds, some of which are in human clinical trials.


Asunto(s)
Interacciones Huésped-Parásitos , Malaria Falciparum , Plasmodium falciparum/crecimiento & desarrollo , Inmunidad Adaptativa , Animales , Antimaláricos/uso terapéutico , Control de Enfermedades Transmisibles/métodos , Culicidae/parasitología , Erradicación de la Enfermedad/métodos , Resistencia a Medicamentos , Eritrocitos/parasitología , Salud Global , Interacciones Huésped-Parásitos/inmunología , Humanos , Estadios del Ciclo de Vida , Hígado/parasitología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Merozoítos/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Esporozoítos/crecimiento & desarrollo , Vacunas Sintéticas/inmunología
2.
Mol Microbiol ; 117(3): 670-681, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34816514

RESUMEN

Infectious diseases remain a major burden to global health. Despite the implementation of successful vaccination campaigns and efficient drugs, the increasing emergence of pathogenic vaccine or treatment resistance demands novel therapeutic strategies. The development of traditional therapies using small-molecule drugs is based on modulating protein function and activity through the occupation of active sites such as enzyme inhibition or ligand-receptor binding. These prerequisites result in the majority of host and pathogenic disease-relevant, nonenzymatic and structural proteins being labeled "undruggable." Targeted protein degradation (TPD) emerged as a powerful strategy to eliminate proteins of interest including those of the undruggable variety. Proteolysis-targeting chimeras (PROTACs) are rationally designed heterobifunctional small molecules that exploit the cellular ubiquitin-proteasome system to specifically mediate the highly selective and effective degradation of target proteins. PROTACs have shown remarkable results in the degradation of various cancer-associated proteins, and several candidates are already in clinical development. Significantly, PROTAC-mediated TPD holds great potential for targeting and modulating pathogenic proteins, especially in the face of increasing drug resistance to the best-in-class treatments. In this review, we discuss advances in the development of TPD in the context of targeting the host-pathogen interface and speculate on their potential use to combat viral, bacterial, and parasitic infection.


Asunto(s)
Complejo de la Endopetidasa Proteasomal , Ubiquitina-Proteína Ligasas , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas/metabolismo , Proteolisis , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
3.
PLoS Biol ; 12(7): e1001897, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24983235

RESUMEN

The malaria parasite Plasmodium falciparum exports several hundred proteins into the infected erythrocyte that are involved in cellular remodeling and severe virulence. The export mechanism involves the Plasmodium export element (PEXEL), which is a cleavage site for the parasite protease, Plasmepsin V (PMV). The PMV gene is refractory to deletion, suggesting it is essential, but definitive proof is lacking. Here, we generated a PEXEL-mimetic inhibitor that potently blocks the activity of PMV isolated from P. falciparum and Plasmodium vivax. Assessment of PMV activity in P. falciparum revealed PEXEL cleavage occurs cotranslationaly, similar to signal peptidase. Treatment of P. falciparum-infected erythrocytes with the inhibitor caused dose-dependent inhibition of PEXEL processing as well as protein export, including impaired display of the major virulence adhesin, PfEMP1, on the erythrocyte surface, and cytoadherence. The inhibitor killed parasites at the trophozoite stage and knockdown of PMV enhanced sensitivity to the inhibitor, while overexpression of PMV increased resistance. This provides the first direct evidence that PMV activity is essential for protein export in Plasmodium spp. and for parasite survival in human erythrocytes and validates PMV as an antimalarial drug target.


Asunto(s)
Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Proteasas de Ácido Aspártico/antagonistas & inhibidores , Oligopéptidos/farmacología , Proteínas Protozoarias/antagonistas & inhibidores , Sulfonamidas/farmacología , Retículo Endoplásmico/metabolismo , Eritrocitos/parasitología , Humanos , Transporte de Proteínas/efectos de los fármacos , Proteínas Protozoarias/metabolismo
4.
Traffic ; 14(5): 532-50, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23387285

RESUMEN

Plasmodium falciparum exports several hundred effector proteins that remodel the host erythrocyte and enable parasites to acquire nutrients, sequester in the circulation and evade immune responses. The majority of exported proteins contain the Plasmodium export element (PEXEL; RxLxE/Q/D) in their N-terminus, which is proteolytically cleaved in the parasite endoplasmic reticulum by Plasmepsin V, and is necessary for export. Several exported proteins lack a PEXEL or contain noncanonical motifs. Here, we assessed whether Plasmepsin V could process the N-termini of diverse protein families in P. falciparum. We show that Plasmepsin V cleaves N-terminal sequences from RIFIN, STEVOR and RESA multigene families, the latter of which contain a relaxed PEXEL (RxLxxE). However, Plasmepsin V does not cleave the N-terminal sequence of the major exported virulence factor erythrocyte membrane protein 1 (PfEMP1) or the PEXEL-negative exported proteins SBP-1 or REX-2. We probed the substrate specificity of Plasmepsin V and determined that lysine at the PEXEL P3 position, which is present in PfEMP1 and other putatively exported proteins, blocks Plasmepsin V activity. Furthermore, isoleucine at position P1 also blocked Plasmepsin V activity. The specificity of Plasmepsin V is therefore exquisitely confined and we have used this novel information to redefine the predicted P. falciparum PEXEL exportome.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Eritrocitos/parasitología , Proteínas de la Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Secuencias de Aminoácidos , Antígenos de Protozoos/metabolismo , Proteínas Portadoras/metabolismo , Cromatografía Líquida de Alta Presión , Biología Computacional , Retículo Endoplásmico/metabolismo , Eritrocitos/citología , Humanos , Estructura Terciaria de Proteína , Programas Informáticos , Fracciones Subcelulares , Factores de Virulencia/metabolismo
5.
J Biol Chem ; 289(37): 25655-69, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25074930

RESUMEN

Plasmodium falciparum is the causative agent of the most severe form of malaria in humans. The merozoite, an extracellular stage of the parasite lifecycle, invades erythrocytes in which they develop. The most abundant protein on the surface of merozoites is merozoite surface protein 1 (MSP1), which consists of four processed fragments. Studies indicate that MSP1 interacts with other peripheral merozoite surface proteins to form a large complex. Successful invasion of merozoites into host erythrocytes is dependent on this protein complex; however, the identity of all components and its function remain largely unknown. We have shown that the peripheral merozoite surface proteins MSPDBL1 and MSPDBL2 are part of the large MSP1 complex. Using surface plasmon resonance, we determined the binding affinities of MSPDBL1 and MSPDBL2 to MSP1 to be in the range of 2-4 × 10(-7) m. Both proteins bound to three of the four proteolytically cleaved fragments of MSP1 (p42, p38, and p83). In addition, MSPDBL1 and MSPDBL2, but not MSP1, bound directly to human erythrocytes. This demonstrates that the MSP1 complex acts as a platform for display of MSPDBL1 and MSPDBL2 on the merozoite surface for binding to receptors on the erythrocyte and invasion.


Asunto(s)
Malaria/metabolismo , Proteína 1 de Superficie de Merozoito/metabolismo , Merozoítos/química , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Eritrocitos/química , Eritrocitos/parasitología , Humanos , Malaria/parasitología , Malaria/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteína 1 de Superficie de Merozoito/química , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Plasmodium falciparum/patogenicidad , Unión Proteica
6.
Malar J ; 14: 280, 2015 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-26187846

RESUMEN

BACKGROUND: Gliding motility in Plasmodium parasites, the aetiological agents of malaria disease, is mediated by an actomyosin motor anchored in the outer pellicle of the motile cell. Effective motility is dependent on a parasite myosin motor and turnover of dynamic parasite actin filaments. To date, however, the basis for directional motility is not known. Whilst myosin is very likely orientated as a result of its anchorage within the parasite, how actin filaments are orientated to facilitate directional force generation remains unexplained. In addition, recent evidence has questioned the linkage between actin filaments and secreted surface antigens leaving the way by which motor force is transmitted to the extracellular milieu unknown. Malaria parasites possess a markedly reduced repertoire of actin regulators, among which few are predicted to interact with filamentous (F)-actin directly. One of these, PF3D7_1251200, shows strong homology to the coronin family of actin-filament binding proteins, herein referred to as PfCoronin. METHODS: Here the N terminal beta propeller domain of PfCoronin (PfCor-N) was expressed to assess its ability to bind and bundle pre-formed actin filaments by sedimentation assay, total internal reflection fluorescence (TIRF) microscopy and confocal imaging as well as to explore its ability to bind phospholipids. In parallel a tagged PfCoronin line in Plasmodium falciparum was generated to determine the cellular localization of the protein during asexual parasite development and blood-stage merozoite invasion. RESULTS: A combination of biochemical approaches demonstrated that the N-terminal beta-propeller domain of PfCoronin is capable of binding F-actin and facilitating formation of parallel filament bundles. In parasites, PfCoronin is expressed late in the asexual lifecycle and localizes to the pellicle region of invasive merozoites before and during erythrocyte entry. PfCoronin also associates strongly with membranes within the cell, likely mediated by interactions with phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) at the plasma membrane. CONCLUSIONS: These data suggest PfCoronin may fulfil a key role as the critical determinant of actin filament organization in the Plasmodium cell. This raises the possibility that macro-molecular organization of actin mediates directional motility in gliding parasites.


Asunto(s)
Citoesqueleto de Actina/química , Proteínas de Microfilamentos/química , Plasmodium falciparum/química , Plasmodium falciparum/fisiología , Proteínas Protozoarias/química , Citoesqueleto de Actina/metabolismo , Animales , Eritrocitos/parasitología , Humanos , Malaria Falciparum/parasitología , Proteínas de Microfilamentos/metabolismo , Modelos Moleculares , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Conejos
7.
Traffic ; 13(11): 1457-65, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22844982

RESUMEN

The establishment of parasite infection within the human erythrocyte is an essential stage in the development of malaria disease. As such, significant interest has focused on the mechanics that underpin invasion and on characterization of parasite molecules involved. Previous evidence has implicated a presenilin-like signal peptide peptidase (SPP) from the most virulent human malaria parasite, Plasmodium falciparum, in the process of invasion where it has been proposed to function in the cleavage of the erythrocyte cytoskeletal protein Band 3. The role of a traditionally endoplasmic reticulum (ER) protease in the process of red blood cell invasion is unexpected. Here, using a combination of molecular, cellular and chemical approaches we provide evidence that PfSPP is, instead, a bona fide ER-resident peptidase that remains intracellular throughout the invasion process. Furthermore, SPP-specific drug inhibition has no effect on erythrocyte invasion whilst having low micromolar potency against intra-erythrocytic development. Contrary to previous reports, these results show that PfSPP plays no role in erythrocyte invasion. Nonetheless, PfSPP clearly represents a potential chemotherapeutic target to block parasite growth, supporting ongoing efforts to develop antimalarial-targeting protein maturation and trafficking during intra-erythrocytic development.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Retículo Endoplásmico/enzimología , Plasmodium falciparum/enzimología , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Eritrocitos/enzimología , Eritrocitos/parasitología , Humanos , Merozoítos/enzimología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad , Inhibidores de Proteasas/farmacología
8.
Proc Natl Acad Sci U S A ; 108(24): 9869-74, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21628589

RESUMEN

Malaria parasite cell motility is a process that is dependent on the dynamic turnover of parasite-derived actin filaments. Despite its central role, actin's polymerization state is controlled by a set of identifiable regulators that is markedly reduced compared with those of other eukaryotic cells. In Plasmodium falciparum, the most virulent species that affects humans, this minimal repertoire includes two members of the actin-depolymerizing factor/cofilin (AC) family of proteins, P. falciparum actin-depolymerizing factor 1 (PfADF1) and P. falciparum actin-depolymerizing factor 2. This essential class of actin regulator is involved in the control of filament dynamics at multiple levels, from monomer binding through to filament depolymerization and severing. Previous biochemical analyses have suggested that PfADF1 sequesters monomeric actin but, unlike most eukaryotic counterparts, has limited potential to bind or depolymerize filaments. The molecular basis for these unusual properties and implications for parasite cell motility have not been established. Here we present the crystal structure of an apicomplexan AC protein, PfADF1. We show that PfADF1 lacks critical residues previously implicated as essential for AC-mediated actin filament binding and disassembly, having a substantially reduced filament-binding loop and C-terminal α4 helix. Despite this divergence in structure, we demonstrate that PfADF1 is capable of efficient actin filament severing. Furthermore, this severing occurs despite PfADF1's low binding affinity for filaments. Comparative structural analysis along with biochemical and microscopy evidence establishes that severing is reliant on the availability of an exposed basic residue in the filament-binding loop, a conserved minimal requirement that defines AC-mediated filament disassembly across eukaryotic cells.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Actinas/metabolismo , Proteínas Protozoarias/metabolismo , Factores Despolimerizantes de la Actina/química , Factores Despolimerizantes de la Actina/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Cristalografía por Rayos X , Humanos , Immunoblotting , Malaria/parasitología , Microscopía Fluorescente/métodos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Homología de Secuencia de Aminoácido
9.
Commun Biol ; 6(1): 861, 2023 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-37596377

RESUMEN

The malaria parasite uses actin-based mechanisms throughout its lifecycle to control a range of biological processes including intracellular trafficking, gene regulation, parasite motility and invasion. In this work we assign functions to the Plasmodium falciparum formins 1 and 2 (FRM1 and FRM2) proteins in asexual and sexual blood stage development. We show that FRM1 is essential for merozoite invasion and FRM2 is required for efficient cell division. We also observed divergent functions for FRM1 and FRM2 in gametocyte development. Conditional deletion of FRM1 leads to a delay in gametocyte stage progression. We show that FRM2 controls the actin and microtubule cytoskeletons in developing gametocytes, with premature removal of the protein resulting in a loss of transmissible stage V gametocytes. Lastly, we show that targeting formin proteins with the small molecule inhibitor of formin homology domain 2 (SMIFH2) leads to a multistage block in asexual and sexual stage parasite development.


Asunto(s)
Actinas , Plasmodium falciparum , Actinas/genética , Forminas , Plasmodium falciparum/genética , División Celular , Citoesqueleto
10.
Nat Commun ; 12(1): 3620, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34131147

RESUMEN

Host membrane remodeling is indispensable for viruses, bacteria, and parasites, to subvert the membrane barrier and obtain entry into cells. The malaria parasite Plasmodium spp. induces biophysical and molecular changes to the erythrocyte membrane through the ordered secretion of its apical organelles. To understand this process and address the debate regarding how the parasitophorous vacuole membrane (PVM) is formed, we developed an approach using lattice light-sheet microscopy, which enables the parasite interaction with the host cell membrane to be tracked and characterized during invasion. Our results show that the PVM is predominantly formed from the erythrocyte membrane, which undergoes biophysical changes as it is remodeled across all stages of invasion, from pre-invasion through to PVM sealing. This approach enables a functional interrogation of parasite-derived lipids and proteins in PVM biogenesis and echinocytosis during Plasmodium falciparum invasion and promises to yield mechanistic insights regarding how this is more generally orchestrated by other intracellular pathogens.


Asunto(s)
Membrana Eritrocítica/parasitología , Eritrocitos/parasitología , Tomografía Computarizada Cuatridimensional/métodos , Interacciones Huésped-Parásitos/fisiología , Malaria/parasitología , Vacuolas/metabolismo , Animales , Membrana Eritrocítica/metabolismo , Humanos , Merozoítos , Parásitos , Plasmodium/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo
11.
Nat Commun ; 11(1): 1780, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286267

RESUMEN

A promising new compound class for treating human malaria is the imidazolopiperazines (IZP) class. IZP compounds KAF156 (Ganaplacide) and GNF179 are effective against Plasmodium symptomatic asexual blood-stage infections, and are able to prevent transmission and block infection in animal models. But despite the identification of resistance mechanisms in P. falciparum, the mode of action of IZPs remains unknown. To investigate, we here combine in vitro evolution and genome analysis in Saccharomyces cerevisiae with molecular, metabolomic, and chemogenomic methods in P. falciparum. Our findings reveal that IZP-resistant S. cerevisiae clones carry mutations in genes involved in Endoplasmic Reticulum (ER)-based lipid homeostasis and autophagy. In Plasmodium, IZPs inhibit protein trafficking, block the establishment of new permeation pathways, and cause ER expansion. Our data highlight a mechanism for blocking parasite development that is distinct from those of standard compounds used to treat malaria, and demonstrate the potential of IZPs for studying ER-dependent protein processing.


Asunto(s)
Antimaláricos/farmacología , Plasmodium falciparum/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Concentración 50 Inhibidora , Espectrometría de Masas , Proteínas Protozoarias/metabolismo , Pirazoles/farmacología , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/metabolismo , Vías Secretoras/efectos de los fármacos
12.
mBio ; 10(5)2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31594816

RESUMEN

Understanding the mechanisms behind host cell invasion by Plasmodium falciparum remains a major hurdle to developing antimalarial therapeutics that target the asexual cycle and the symptomatic stage of malaria. Host cell entry is enabled by a multitude of precisely timed and tightly regulated receptor-ligand interactions. Cyclic nucleotide signaling has been implicated in regulating parasite invasion, and an important downstream effector of the cAMP-signaling pathway is protein kinase A (PKA), a cAMP-dependent protein kinase. There is increasing evidence that P. falciparum PKA (PfPKA) is responsible for phosphorylation of the cytoplasmic domain of P. falciparum apical membrane antigen 1 (PfAMA1) at Ser610, a cAMP-dependent event that is crucial for successful parasite invasion. In the present study, CRISPR-Cas9 and conditional gene deletion (dimerizable cre) technologies were implemented to generate a P. falciparum parasite line in which expression of the catalytic subunit of PfPKA (PfPKAc) is under conditional control, demonstrating highly efficient dimerizable Cre recombinase (DiCre)-mediated gene excision and complete knockdown of protein expression. Parasites lacking PfPKAc show severely reduced growth after one intraerythrocytic growth cycle and are deficient in host cell invasion, as highlighted by live-imaging experiments. Furthermore, PfPKAc-deficient parasites are unable to phosphorylate PfAMA1 at Ser610. This work not only identifies an essential role for PfPKAc in the P. falciparum asexual life cycle but also confirms that PfPKAc is the kinase responsible for phosphorylating PfAMA1 Ser610.IMPORTANCE Malaria continues to present a major global health burden, particularly in low-resource countries. Plasmodium falciparum, the parasite responsible for the most severe form of malaria, causes disease through rapid and repeated rounds of invasion and replication within red blood cells. Invasion into red blood cells is essential for P. falciparum survival, and the molecular events mediating this process have gained much attention as potential therapeutic targets. With no effective vaccine available, and with the emergence of resistance to antimalarials, there is an urgent need for the development of new therapeutics. Our research has used genetic techniques to provide evidence of an essential protein kinase involved in P. falciparum invasion. Our work adds to the current understanding of parasite signaling processes required for invasion, highlighting PKA as a potential drug target to inhibit invasion for the treatment of malaria.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Endocitosis , Eritrocitos/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Proteínas Protozoarias/metabolismo , Antígenos de Protozoos/metabolismo , Dominio Catalítico , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Humanos , Proteínas de la Membrana/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional
13.
mBio ; 10(6)2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31822583

RESUMEN

Members of the haloacid dehalogenase (HAD) family of metabolite phosphatases play an important role in regulating multiple pathways in Plasmodium falciparum central carbon metabolism. We show that the P. falciparum HAD protein, phosphoglycolate phosphatase (PGP), regulates glycolysis and pentose pathway flux in asexual blood stages via detoxifying the damaged metabolite 4-phosphoerythronate (4-PE). Disruption of the P. falciparumpgp gene caused accumulation of two previously uncharacterized metabolites, 2-phospholactate and 4-PE. 4-PE is a putative side product of the glycolytic enzyme, glyceraldehyde-3-phosphate dehydrogenase, and its accumulation inhibits the pentose phosphate pathway enzyme, 6-phosphogluconate dehydrogenase (6-PGD). Inhibition of 6-PGD by 4-PE leads to an unexpected feedback response that includes increased flux into the pentose phosphate pathway as a result of partial inhibition of upper glycolysis, with concomitant increased sensitivity to antimalarials that target pathways downstream of glycolysis. These results highlight the role of metabolite detoxification in regulating central carbon metabolism and drug sensitivity of the malaria parasite.IMPORTANCE The malaria parasite has a voracious appetite, requiring large amounts of glucose and nutrients for its rapid growth and proliferation inside human red blood cells. The host cell is resource rich, but this is a double-edged sword; nutrient excess can lead to undesirable metabolic reactions and harmful by-products. Here, we demonstrate that the parasite possesses a metabolite repair enzyme (PGP) that suppresses harmful metabolic by-products (via substrate dephosphorylation) and allows the parasite to maintain central carbon metabolism. Loss of PGP leads to the accumulation of two damaged metabolites and causes a domino effect of metabolic dysregulation. Accumulation of one damaged metabolite inhibits an essential enzyme in the pentose phosphate pathway, leading to substrate accumulation and secondary inhibition of glycolysis. This work highlights how the parasite coordinates metabolic flux by eliminating harmful metabolic by-products to ensure rapid proliferation in its resource-rich niche.


Asunto(s)
Antimaláricos/farmacología , Carbono/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Fosfomicina/análogos & derivados , Monoéster Fosfórico Hidrolasas/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Fosfomicina/farmacología , Glucólisis/efectos de los fármacos , Humanos , Lactatos/farmacología , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/metabolismo , Azúcares Ácidos/farmacología
14.
Nat Microbiol ; 3(9): 1010-1022, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30127496

RESUMEN

Plasmodium falciparum exports hundreds of virulence proteins within infected erythrocytes, a process that requires cleavage of a pentameric motif called Plasmodium export element or vacuolar transport signal by the endoplasmic reticulum (ER)-resident protease plasmepsin V. We identified plasmepsin V-binding proteins that form a unique interactome required for the translocation of effector cargo into the parasite ER. These interactions are functionally distinct from the Sec61-signal peptidase complex required for the translocation of proteins destined for the classical secretory pathway. This interactome does not involve the signal peptidase (SPC21) and consists of PfSec61, PfSPC25, plasmepsin V and PfSec62, which is an essential component of the post-translational ER translocon. Together, they form a distinct portal for the recognition and translocation of a large subset of Plasmodium export element effector proteins into the ER, thereby remodelling the infected erythrocyte that is required for parasite survival and pathogenesis.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Retículo Endoplásmico/metabolismo , Eritrocitos/metabolismo , Plasmodium falciparum/metabolismo , Canales de Translocación SEC/metabolismo , Ácido Aspártico Endopeptidasas/genética , Transporte Biológico/genética , Transporte Biológico/fisiología , Membrana Celular/fisiología , Malaria Falciparum/patología , Plasmodium falciparum/patogenicidad , Canales de Translocación SEC/genética , Factores de Virulencia
15.
Nat Microbiol ; 2: 17031, 2017 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-28288098

RESUMEN

Malaria control is heavily dependent on chemotherapeutic agents for disease prevention and drug treatment. Defining the mechanism of action for licensed drugs, for which no target is characterized, is critical to the development of their second-generation derivatives to improve drug potency towards inhibition of their molecular targets. Mefloquine is a widely used antimalarial without a known mode of action. Here, we demonstrate that mefloquine is a protein synthesis inhibitor. We solved a 3.2 Šcryo-electron microscopy structure of the Plasmodium falciparum 80S ribosome with the (+)-mefloquine enantiomer bound to the ribosome GTPase-associated centre. Mutagenesis of mefloquine-binding residues generates parasites with increased resistance, confirming the parasite-killing mechanism. Furthermore, structure-guided derivatives with an altered piperidine group, predicted to improve binding, show enhanced parasiticidal effect. These data reveal one possible mode of action for mefloquine and demonstrate the vast potential of cryo-electron microscopy to guide the development of mefloquine derivatives to inhibit parasite protein synthesis.


Asunto(s)
Antimaláricos/farmacología , Mefloquina/farmacología , Plasmodium falciparum/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Ribosomas/efectos de los fármacos
16.
Cell Host Microbe ; 20(1): 60-71, 2016 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-27374406

RESUMEN

Plasmodium falciparum parasites in the merozoite stage invade human erythrocytes and cause malaria. Invasion requires multiple interactions between merozoite ligands and erythrocyte receptors. P. falciparum reticulocyte binding homolog 5 (PfRh5) forms a complex with the PfRh5-interacting protein (PfRipr) and Cysteine-rich protective antigen (CyRPA) and binds erythrocytes via the host receptor basigin. However, the specific role that PfRipr and CyRPA play during invasion is unclear. Using P. falciparum lines conditionally expressing PfRipr and CyRPA, we show that loss of PfRipr or CyRPA function blocks growth due to the inability of merozoites to invade erythrocytes. Super-resolution microscopy revealed that PfRipr, CyRPA, and PfRh5 colocalize at the junction between merozoites and erythrocytes during invasion. PfRipr, CyRPA, and PfRipr/CyRPA/PfRh5-basigin complex is required for triggering the Ca(2+) release and establishing the tight junction. Together, these results establish that the PfRh5/PfRipr/CyRPA complex is essential in the sequential molecular events leading to parasite invasion of human erythrocytes.


Asunto(s)
Antígenos de Protozoos/metabolismo , Proteínas Portadoras/metabolismo , Endocitosis , Eritrocitos/parasitología , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Basigina/metabolismo , Calcio/metabolismo , Cationes Bivalentes/metabolismo , Técnicas de Silenciamiento del Gen , Interacciones Huésped-Patógeno , Humanos , Microscopía , Modelos Biológicos , Unión Proteica , Multimerización de Proteína
17.
Science ; 367(6473): 22-23, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31896702
18.
PLoS One ; 7(9): e46160, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23049965

RESUMEN

Host cell infection by apicomplexan parasites plays an essential role in lifecycle progression for these obligate intracellular pathogens. For most species, including the etiological agents of malaria and toxoplasmosis, infection requires active host-cell invasion dependent on formation of a tight junction - the organising interface between parasite and host cell during entry. Formation of this structure is not, however, shared across all Apicomplexa or indeed all parasite lifecycle stages. Here, using an in silico integrative genomic search and endogenous gene-tagging strategy, we sought to characterise proteins that function specifically during junction-dependent invasion, a class of proteins we term invasins to distinguish them from adhesins that function in species specific host-cell recognition. High-definition imaging of tagged Plasmodium falciparum invasins localised proteins to multiple cellular compartments of the blood stage merozoite. This includes several that localise to distinct subcompartments within the rhoptries. While originating from the same organelle, however, each has very different dynamics during invasion. Apical Sushi Protein and Rhoptry Neck protein 2 release early, following the junction, whilst a novel rhoptry protein PFF0645c releases only after invasion is complete. This supports the idea that organisation of proteins within a secretory organelle determines the order and destination of protein secretion and provides a localisation-based classification strategy for predicting invasin function during apicomplexan parasite invasion.


Asunto(s)
Eritrocitos/parasitología , Malaria/parasitología , Orgánulos/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Western Blotting , Técnica del Anticuerpo Fluorescente , Interacciones Huésped-Parásitos , Humanos , Microscopía Inmunoelectrónica , Orgánulos/ultraestructura
19.
PLoS One ; 7(2): e32188, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22389687

RESUMEN

Actin dynamics have been implicated in a variety of developmental processes during the malaria parasite lifecycle. Parasite motility, in particular, is thought to critically depend on an actomyosin motor located in the outer pellicle of the parasite cell. Efforts to understand the diverse roles actin plays have, however, been hampered by an inability to detect microfilaments under native conditions. To visualise the spatial dynamics of actin we generated a parasite-specific actin antibody that shows preferential recognition of filamentous actin and applied this tool to different lifecycle stages (merozoites, sporozoites and ookinetes) of the human and mouse malaria parasite species Plasmodium falciparum and P. berghei along with tachyzoites from the related apicomplexan parasite Toxoplasma gondii. Actin filament distribution was found associated with three core compartments: the nuclear periphery, pellicular membranes of motile or invasive parasite forms and in a ring-like distribution at the tight junction during merozoite invasion of erythrocytes in both human and mouse malaria parasites. Localisation at the nuclear periphery is consistent with an emerging role of actin in facilitating parasite gene regulation. During invasion, we show that the actin ring at the parasite-host cell tight junction is dependent on dynamic filament turnover. Super-resolution imaging places this ring posterior to, and not concentric with, the junction marker rhoptry neck protein 4. This implies motor force relies on the engagement of dynamic microfilaments at zones of traction, though not necessarily directly through receptor-ligand interactions at sites of adhesion during invasion. Combined, these observations extend current understanding of the diverse roles actin plays in malaria parasite development and apicomplexan cell motility, in particular refining understanding on the linkage of the internal parasite gliding motor with the extra-cellular milieu.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Malaria/parasitología , Proteínas Protozoarias/metabolismo , Citoesqueleto de Actina/química , Animales , Humanos , Estadios del Ciclo de Vida/fisiología , Merozoítos/metabolismo , Ratones , Plasmodium berghei/metabolismo , Plasmodium falciparum/metabolismo , Estructura Secundaria de Proteína , Esporozoítos/metabolismo
20.
Cell Host Microbe ; 9(1): 9-20, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21238943

RESUMEN

Erythrocyte invasion by the merozoite is an obligatory stage in Plasmodium parasite infection and essential to malaria disease progression. Attempts to study this process have been hindered by the poor invasion synchrony of merozoites from the only in vitro culture-adapted human malaria parasite, Plasmodium falciparum. Using fluorescence, three-dimensional structured illumination, and immunoelectron microscopy of filtered merozoites, we analyze cellular and molecular events underlying each discrete step of invasion. Monitoring the dynamics of these events revealed that commitment to the process is mediated through merozoite attachment to the erythrocyte, triggering all subsequent invasion events, which then proceed without obvious checkpoints. Instead, coordination of the invasion process involves formation of the merozoite-erythrocyte tight junction, which acts as a nexus for rhoptry secretion, surface-protein shedding, and actomyosin motor activation. The ability to break down each molecular step allows us to propose a comprehensive model for the molecular basis of parasite invasion.


Asunto(s)
Eritrocitos/parasitología , Eritrocitos/ultraestructura , Malaria Falciparum/patología , Malaria Falciparum/parasitología , Plasmodium falciparum/patogenicidad , Plasmodium falciparum/ultraestructura , Adhesión Celular , Humanos , Imagenología Tridimensional , Merozoítos/fisiología , Merozoítos/ultraestructura , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Modelos Biológicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA