Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
ACS Infect Dis ; 9(9): 1695-1710, 2023 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-37639221

RESUMO

With the resistance increasing to current antimalarial medicines, there is an urgent need to discover new drug targets and to develop new medicines against these targets. We therefore screened the Open Global Health Library of Merck KGaA, Darmstadt, Germany, of 250 compounds against the asexual blood stage of the deadliest malarial parasite Plasmodium falciparum, from which eight inhibitors with low micromolar potency were found. Due to its combined potencies against parasite growth and inhibition of red blood cell invasion, the pyridyl-furan compound OGHL250 was prioritized for further optimization. The potency of the series lead compound (WEHI-518) was improved 250-fold to low nanomolar levels against parasite blood-stage growth. Parasites selected for resistance to a related compound, MMV396797, were also resistant to WEHI-518 as well as KDU731, an inhibitor of the phosphatidylinositol kinase PfPI4KIIIB, suggesting that this kinase is the target of the pyridyl-furan series. Inhibition of PfPI4KIIIB blocks multiple stages of the parasite's life cycle and other potent inhibitors are currently under preclinical development. MMV396797-resistant parasites possess an E1316D mutation in PfPKI4IIIB that clusters with known resistance mutations of other inhibitors of the kinase. Building upon earlier studies that showed that PfPI4KIIIB inhibitors block the development of the invasive merozoite parasite stage, we show that members of the pyridyl-furan series also block invasion and/or the conversion of merozoites into ring-stage intracellular parasites through inhibition of protein secretion and export into red blood cells.


Assuntos
Parasitos , Animais , Plasmodium falciparum/genética , Saúde Global , Eritrócitos , Transporte Proteico , Furanos
2.
PLoS Pathog ; 19(7): e1011006, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37523385

RESUMO

A key element of Plasmodium biology and pathogenesis is the trafficking of ~10% of the parasite proteome into the host red blood cell (RBC) it infects. To cross the parasite-encasing parasitophorous vacuole membrane, exported proteins utilise a channel-forming protein complex termed the Plasmodium translocon of exported proteins (PTEX). PTEX is obligatory for parasite survival, both in vitro and in vivo, suggesting that at least some exported proteins have essential metabolic functions. However, to date only one essential PTEX-dependent process, the new permeability pathways, has been described. To identify other essential PTEX-dependant proteins/processes, we conditionally knocked down the expression of one of its core components, PTEX150, and examined which pathways were affected. Surprisingly, the food vacuole mediated process of haemoglobin (Hb) digestion was substantially perturbed by PTEX150 knockdown. Using a range of transgenic parasite lines and approaches, we show that two major Hb proteases; falcipain 2a and plasmepsin II, interact with PTEX core components, implicating the translocon in the trafficking of Hb proteases. We propose a model where these proteases are translocated into the PV via PTEX in order to reach the cytostome, located at the parasite periphery, prior to food vacuole entry. This work offers a second mechanistic explanation for why PTEX function is essential for growth of the parasite within its host RBC.


Assuntos
Parasitos , Plasmodium falciparum , Animais , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Vacúolos/metabolismo , Transporte Proteico , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Eritrócitos/parasitologia , Parasitos/metabolismo , Peptídeo Hidrolases/metabolismo
3.
Traffic ; 23(9): 442-461, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36040075

RESUMO

Plasmodium falciparum parasites which cause malaria, traffic hundreds of proteins into the red blood cells (RBCs) they infect. These exported proteins remodel their RBCs enabling host immune evasion through processes such as cytoadherence that greatly assist parasite survival. As resistance to all current antimalarial compounds is rising new compounds need to be identified and those that could inhibit parasite protein secretion and export would both rapidly reduce parasite virulence and ultimately lead to parasite death. To identify compounds that inhibit protein export we used transgenic parasites expressing an exported nanoluciferase reporter to screen the Medicines for Malaria Venture Malaria Box of 400 antimalarial compounds with mostly unknown targets. The most potent inhibitor identified in this screen was MMV396797 whose application led to export inhibition of both the reporter and endogenous exported proteins. MMV396797 mediated blockage of protein export and slowed the rigidification and cytoadherence of infected RBCs-modifications which are both mediated by parasite-derived exported proteins. Overall, we have identified a new protein export inhibitor in P. falciparum whose target though unknown, could be developed into a future antimalarial that rapidly inhibits parasite virulence before eliminating parasites from the host.


Assuntos
Antimaláricos , Malária Falciparum , Malária , Parasitos , Animais , Antimaláricos/metabolismo , Antimaláricos/farmacologia , Antimaláricos/uso terapêutico , Eritrócitos/parasitologia , Humanos , Malária Falciparum/tratamento farmacológico , Malária Falciparum/parasitologia , Parasitos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo
4.
Mol Microbiol ; 117(5): 1245-1262, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35403274

RESUMO

Infection with Plasmodium falciparum parasites results in approximately 627,000 deaths from malaria annually. Key to the parasite's success is their ability to invade and subsequently grow within human erythrocytes. Parasite proteins involved in parasite invasion and proliferation are therefore intrinsically of great interest, as targeting these proteins could provide novel means of therapeutic intervention. One such protein is P113 which has been reported to be both an invasion protein and an intracellular protein located within the parasitophorous vacuole (PV). The PV is delimited by a membrane (PVM) across which a plethora of parasite-specific proteins are exported via the Plasmodium Translocon of Exported proteins (PTEX) into the erythrocyte to enact various immune evasion functions. To better understand the role of P113 we isolated its binding partners from in vitro cultures of P. falciparum. We detected interactions with the protein export machinery (PTEX and exported protein-interacting complex) and a variety of proteins that either transit through the PV or reside on the parasite plasma membrane. Genetic knockdown or partial deletion of P113 did not significantly reduce parasite growth or protein export but did disrupt the morphology of the PVM, suggesting that P113 may play a role in maintaining normal PVM architecture.


Assuntos
Malária Falciparum , Parasitos , Animais , Eritrócitos/parasitologia , Humanos , Malária Falciparum/parasitologia , Parasitos/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Transporte Proteico/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Vacúolos/metabolismo
5.
PLoS Pathog ; 18(2): e1009977, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35192672

RESUMO

Plasmodium falciparum exports ~10% of its proteome into its host erythrocyte to modify the host cell's physiology. The Plasmodium export element (PEXEL) motif contained within the N-terminus of most exported proteins directs the trafficking of those proteins into the erythrocyte. To reach the host cell, the PEXEL motif of exported proteins is processed by the endoplasmic reticulum (ER) resident aspartyl protease plasmepsin V. Then, following secretion into the parasite-encasing parasitophorous vacuole, the mature exported protein must be unfolded and translocated across the parasitophorous vacuole membrane by the Plasmodium translocon of exported proteins (PTEX). PTEX is a protein-conducting channel consisting of the pore-forming protein EXP2, the protein unfoldase HSP101, and structural component PTEX150. The mechanism of how exported proteins are specifically trafficked from the parasite's ER following PEXEL cleavage to PTEX complexes on the parasitophorous vacuole membrane is currently not understood. Here, we present evidence that EXP2 and PTEX150 form a stable subcomplex that facilitates HSP101 docking. We also demonstrate that HSP101 localises both within the parasitophorous vacuole and within the parasite's ER throughout the ring and trophozoite stage of the parasite, coinciding with the timeframe of protein export. Interestingly, we found that HSP101 can form specific interactions with model PEXEL proteins in the parasite's ER, irrespective of their PEXEL processing status. Collectively, our data suggest that HSP101 recognises and chaperones PEXEL proteins from the ER to the parasitophorous vacuole and given HSP101's specificity for the EXP2-PTEX150 subcomplex, this provides a mechanism for how exported proteins are specifically targeted to PTEX for translocation into the erythrocyte.


Assuntos
Parasitos , Plasmodium falciparum , Animais , Eritrócitos/parasitologia , Parasitos/metabolismo , Plasmodium falciparum/metabolismo , Transporte Proteico/fisiologia , Proteínas de Protozoários/metabolismo
6.
Bioorg Chem ; 117: 105359, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34689083

RESUMO

Malaria is a devastating disease caused by Plasmodium parasites. Emerging resistance against current antimalarial therapeutics has engendered the need to develop antimalarials with novel structural classes. We recently described the identification and initial optimization of the 2-anilino quinazoline antimalarial class. Here, we refine the physicochemical properties of this antimalarial class with the aim to improve aqueous solubility and metabolism and to reduce adverse promiscuity. We show the physicochemical properties of this class are intricately balanced with asexual parasite activity and human cell cytotoxicity. Structural modifications we have implemented improved LipE, aqueous solubility and in vitro metabolism while preserving fast acting P. falciparum asexual stage activity. The lead compounds demonstrated equipotent activity against P. knowlesi parasites and were not predisposed to resistance mechanisms of clinically used antimalarials. The optimized compounds exhibited modest activity against early-stage gametocytes, but no activity against pre-erythrocytic liver parasites. Confoundingly, the refined physicochemical properties installed in the compounds did not engender improved oral efficacy in a P. berghei mouse model of malaria compared to earlier studies on the 2-anilino quinazoline class. This study provides the framework for further development of this antimalarial class.


Assuntos
Compostos de Anilina/química , Compostos de Anilina/farmacologia , Antimaláricos/química , Antimaláricos/farmacologia , Malária/tratamento farmacológico , Plasmodium/efeitos dos fármacos , Quinazolinas/química , Quinazolinas/farmacologia , Aminação , Compostos de Anilina/uso terapêutico , Animais , Antimaláricos/uso terapêutico , Feminino , Humanos , Malária/parasitologia , Camundongos , Plasmodium/fisiologia , Plasmodium falciparum/efeitos dos fármacos , Plasmodium falciparum/fisiologia , Quinazolinas/uso terapêutico
7.
Cell Microbiol ; 23(8): e13332, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33774908

RESUMO

During its intraerythrocytic life cycle, the human malaria parasite Plasmodium falciparum supplements its nutritional requirements by scavenging substrates from the plasma through the new permeability pathways (NPPs) installed in the red blood cell (RBC) membrane. Parasite proteins of the RhopH complex: CLAG3, RhopH2, RhopH3, have been implicated in NPP activity. Here, we studied 13 exported proteins previously hypothesised to interact with RhopH2, to study their potential contribution to the function of NPPs. NPP activity assays revealed that the 13 proteins do not appear to be individually important for NPP function, as conditional knockdown of these proteins had no effect on sorbitol uptake. Intriguingly, reciprocal immunoprecipitation assays showed that five of the 13 proteins interact with all members of the RhopH complex, with PF3D7_1401200 showing the strongest association. Mass spectrometry-based proteomics further identified new protein complexes; a cytoskeletal complex and a Maurer's clefts/J-dot complex, which overall helps clarify protein-protein interactions within the infected RBC (iRBC) and is suggestive of the potential trafficking route of the RhopH complex itself to the RBC membrane.


Assuntos
Parasitos , Plasmodium falciparum , Animais , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Humanos , Parasitos/metabolismo , Plasmodium falciparum/metabolismo , Transporte Proteico , Proteínas de Protozoários/metabolismo
8.
mBio ; 10(5)2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31594816

RESUMO

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.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Endocitose , Eritrócitos/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Proteínas de Protozoários/metabolismo , Antígenos de Protozoários/metabolismo , Domínio Catalítico , Proteínas Quinases Dependentes de AMP Cíclico/genética , Humanos , Proteínas de Membrana/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional
9.
Sci Rep ; 9(1): 10292, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31311978

RESUMO

We developed a novel series of antimalarial compounds based on a 4-cyano-3-methylisoquinoline. Our lead compound MB14 achieved modest inhibition of the growth in vitro of the human malaria parasite, Plasmodium falciparum. To identify its biological target we selected for parasites resistant to MB14. Genome sequencing revealed that all resistant parasites bore a single point S374R mutation in the sodium (Na+) efflux transporter PfATP4. There are many compounds known to inhibit PfATP4 and some are under preclinical development. MB14 was shown to inhibit Na+ dependent ATPase activity in parasite membranes, consistent with the compound targeting PfATP4 directly. PfATP4 inhibitors cause swelling and lysis of infected erythrocytes, attributed to the accumulation of Na+ inside the intracellular parasites and the resultant parasite swelling. We show here that inhibitor-induced lysis of infected erythrocytes is dependent upon the parasite protein RhopH2, a component of the new permeability pathways that are induced by the parasite in the erythrocyte membrane. These pathways mediate the influx of Na+ into the infected erythrocyte and their suppression via RhopH2 knockdown limits the accumulation of Na+ within the parasite hence protecting the infected erythrocyte from lysis. This study reveals a role for the parasite-induced new permeability pathways in the mechanism of action of PfATP4 inhibitors.


Assuntos
Eritrócitos/efeitos dos fármacos , Isoquinolinas/síntese química , Plasmodium falciparum/efeitos dos fármacos , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Membrana Celular/efeitos dos fármacos , Resistência a Medicamentos/efeitos dos fármacos , Eritrócitos/parasitologia , Isoquinolinas/química , Isoquinolinas/farmacologia , Modelos Moleculares , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Mutação Puntual , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Sódio , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/genética , Sequenciamento Completo do Genoma
10.
PLoS Pathog ; 15(5): e1007670, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31121005

RESUMO

To elicit effective invasion and egress from infected cells, obligate intracellular parasites of the phylum Apicomplexa rely on the timely and spatially controlled exocytosis of specialized secretory organelles termed the micronemes. The effector molecules and signaling events underpinning this process are intricate; however, recent advances within the field of Toxoplasma gondii research have facilitated a broader understanding as well as a more integrated view of this complex cascade of events and have unraveled the importance of phosphatidic acid (PA) as a lipid mediator at multiple steps in this process.


Assuntos
Cálcio/metabolismo , GMP Cíclico/metabolismo , Exocitose/fisiologia , Organelas/metabolismo , Ácidos Fosfatídicos/metabolismo , Toxoplasma/fisiologia , Toxoplasmose/parasitologia , Animais , Interações Hospedeiro-Parasita , Humanos , Organelas/parasitologia , Transporte Proteico , Proteínas de Protozoários/metabolismo , Transdução de Sinais
11.
PLoS One ; 13(11): e0204785, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30439948

RESUMO

Malaria parasites remodel their host erythrocytes to gain nutrients and avoid the immune system. Host erythrocytes are modified by hundreds of effector proteins exported from the parasite into the host cell. Protein export is mediated by the PTEX translocon comprising five core components of which EXP2 is considered to form the putative pore that spans the vacuole membrane enveloping the parasite within its erythrocyte. To explore the function and importance of EXP2 for parasite survival in the asexual blood stage of Plasmodium falciparum we inducibly knocked down the expression of EXP2. Reduction in EXP2 expression strongly reduced parasite growth proportional to the degree of protein knockdown and tended to stall development about half way through the asexual cell cycle. Once the knockdown inducer was removed and EXP2 expression restored, parasite growth recovered dependent upon the length and degree of knockdown. To establish EXP2 function and hence the basis for growth reduction, the trafficking of an exported protein was monitored following EXP2 knockdown. This resulted in severe attenuation of protein export and is consistent with EXP2, and PTEX in general, being the conduit for export of proteins into the host compartment.


Assuntos
Proteínas de Transporte , Plasmodium falciparum , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Técnicas de Silenciamento de Genes , Humanos , Estágios do Ciclo de Vida/genética , Plasmodium falciparum/genética , Plasmodium falciparum/crescimento & desenvolvimento , Transporte Proteico/genética , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo
12.
Traffic ; 19(8): 605-623, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29696751

RESUMO

Plasmodium falciparum, which causes malaria, extensively remodels its human host cells, particularly erythrocytes. Remodelling is essential for parasite survival by helping to avoid host immunity and assisting in the uptake of plasma nutrients to fuel rapid growth. Host cell renovation is carried out by hundreds of parasite effector proteins that are exported into the erythrocyte across an enveloping parasitophorous vacuole membrane (PVM). The Plasmodium translocon for exported (PTEX) proteins is thought to span the PVM and provide a channel that unfolds and extrudes proteins across the PVM into the erythrocyte. We show that exported reporter proteins containing mouse dihydrofolate reductase domains that inducibly resist unfolding become trapped at the parasite surface partly colocalizing with PTEX. When cargo is trapped, loop-like extensions appear at the PVM containing both trapped cargo and PTEX protein EXP2, but not additional components HSP101 and PTEX150. Following removal of the block-inducing compound, export of reporter proteins only partly recovers possibly because much of the trapped cargo is spatially segregated in the loop regions away from PTEX. This suggests that parasites have the means to isolate unfoldable cargo proteins from PTEX-containing export zones to avert disruption of protein export that would reduce parasite growth.


Assuntos
Malária Falciparum/parasitologia , Parasitos/metabolismo , Plasmodium falciparum/metabolismo , Transporte Proteico/fisiologia , Proteínas de Protozoários/metabolismo , Animais , Eritrócitos/parasitologia , Humanos , Malária Falciparum/sangue , Camundongos , Vacúolos/parasitologia
13.
Elife ; 62017 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-28252383

RESUMO

Plasmodium falciparum parasites, the causative agents of malaria, modify their host erythrocyte to render them permeable to supplementary nutrient uptake from the plasma and for removal of toxic waste. Here we investigate the contribution of the rhoptry protein RhopH2, in the formation of new permeability pathways (NPPs) in Plasmodium-infected erythrocytes. We show RhopH2 interacts with RhopH1, RhopH3, the erythrocyte cytoskeleton and exported proteins involved in host cell remodeling. Knockdown of RhopH2 expression in cycle one leads to a depletion of essential vitamins and cofactors and decreased de novo synthesis of pyrimidines in cycle two. There is also a significant impact on parasite growth, replication and transition into cycle three. The uptake of solutes that use NPPs to enter erythrocytes is also reduced upon RhopH2 knockdown. These findings provide direct genetic support for the contribution of the RhopH complex in NPP activity and highlight the importance of NPPs to parasite survival.


Assuntos
Eritrócitos/parasitologia , Interações Hospedeiro-Patógeno , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Citoesqueleto/metabolismo , Humanos , Camundongos , Pirimidinas/metabolismo , Vitaminas/metabolismo
14.
J Med Chem ; 60(3): 1171-1188, 2017 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-28080063

RESUMO

Novel antimalarial therapeutics that target multiple stages of the parasite lifecycle are urgently required to tackle the emerging problem of resistance with current drugs. Here, we describe the optimization of the 2-anilino quinazoline class as antimalarial agents. The class, identified from publicly available antimalarial screening data, was optimized to generate lead compounds that possess potent antimalarial activity against P. falciparum parasites comparable to the known antimalarials, chloroquine and mefloquine. During the optimization process, we defined the functionality necessary for activity and improved in vitro metabolism and solubility. The resultant lead compounds possess potent activity against a multidrug resistant strain of P. falciparum and arrest parasites at the ring phase of the asexual stage and also gametocytogensis. Finally, we show that the lead compounds are orally efficacious in a 4 day murine model of malaria disease burden.


Assuntos
Antimaláricos/uso terapêutico , Quinazolinas/uso terapêutico , Administração Oral , Animais , Antimaláricos/administração & dosagem , Antimaláricos/farmacologia , Modelos Animais de Doenças , Camundongos , Plasmodium falciparum/efeitos dos fármacos , Quinazolinas/administração & dosagem , Quinazolinas/farmacologia , Relação Estrutura-Atividade
15.
J Biol Chem ; 291(49): 25375-25386, 2016 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-27738107

RESUMO

The ubiquitous second messenger cAMP mediates signal transduction processes in the malarial parasite that regulate host erythrocyte invasion and the proliferation of merozoites. In Plasmodium falciparum, the central receptor for cAMP is the single regulatory subunit (R) of protein kinase A (PKA). To aid the development of compounds that can selectively dysregulate parasite PKA signaling, we solved the structure of the PKA regulatory subunit in complex with cAMP and a related analogue that displays antimalarial activity, (Sp)-2-Cl-cAMPS. Prior to signaling, PKA-R holds the kinase's catalytic subunit (C) in an inactive state by exerting an allosteric inhibitory effect. When two cAMP molecules bind to PKA-R, they stabilize a structural conformation that facilitates its dissociation, freeing PKA-C to phosphorylate downstream substrates such as apical membrane antigen 1. Although PKA activity was known to be necessary for erythrocytic proliferation, we show that uncontrolled induction of PKA activity using membrane-permeable agonists is equally disruptive to growth.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/química , AMP Cíclico , Plasmodium falciparum/enzimologia , Proteínas de Protozoários/química , Regulação Alostérica , AMP Cíclico/análogos & derivados , AMP Cíclico/química , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas de Protozoários/antagonistas & inibidores , Proteínas de Protozoários/metabolismo
16.
FEBS Lett ; 590(15): 2469-81, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27403735

RESUMO

Lipids are commonly known for the structural roles they play, however, the specific contribution of different lipid classes to wide-ranging signalling pathways is progressively being unravelled. Signalling lipids and their associated effector proteins are emerging as significant contributors to a vast array of effector functions within cells, including essential processes such as membrane fusion and vesicle exocytosis. Many phospholipids have signalling capacity, however, this review will focus on phosphatidic acid (PA) and the enzymes implicated in its production from diacylglycerol (DAG) and phosphatidylcholine (PC): DGK and PLD respectively. PA is a negatively charged, cone-shaped lipid identified as a key mediator in specific membrane fusion and vesicle exocytosis events in a variety of mammalian cells, and has recently been implicated in specialised secretory organelle exocytosis in apicomplexan parasites. This review summarises the recent work implicating a role for PA regulation in exocytosis in various cell types. We will discuss how these signalling events are linked to pathogenesis in the phylum Apicomplexa.


Assuntos
Apicomplexa/genética , Exocitose/genética , Ácidos Fosfatídicos , Fosfatidilcolinas/metabolismo , Apicomplexa/metabolismo , Apicomplexa/patogenicidade , Diglicerídeos/metabolismo , Metabolismo dos Lipídeos/genética , Lipídeos/química , Lipídeos/genética , Transdução de Sinais
17.
Cell Host Microbe ; 19(3): 349-60, 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26962945

RESUMO

The obligate intracellular lifestyle of apicomplexan parasites necessitates an invasive phase underpinned by timely and spatially controlled secretion of apical organelles termed micronemes. In Toxoplasma gondii, extracellular potassium levels and other stimuli trigger a signaling cascade culminating in phosphoinositide-phospholipase C (PLC) activation, which generates the second messengers diacylglycerol (DAG) and IP3 and ultimately results in microneme secretion. Here we show that a delicate balance between DAG and its downstream product, phosphatidic acid (PA), is essential for controlling microneme release. Governing this balance is the apicomplexan-specific DAG-kinase-1, which interconverts PA and DAG, and whose depletion impairs egress and causes parasite death. Additionally, we identify an acylated pleckstrin-homology (PH) domain-containing protein (APH) on the microneme surface that senses PA during microneme secretion and is necessary for microneme exocytosis. As APH is conserved in Apicomplexa, these findings highlight a potentially widely used mechanism in which key lipid mediators regulate microneme exocytosis.


Assuntos
Organelas/efeitos dos fármacos , Organelas/metabolismo , Ácidos Fosfatídicos/metabolismo , Proteínas de Protozoários/metabolismo , Transdução de Sinais , Toxoplasma/fisiologia , Diacilglicerol Quinase/metabolismo , Diglicerídeos/metabolismo
18.
Cell Microbiol ; 18(11): 1551-1569, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27019089

RESUMO

The Plasmodium translocon for exported proteins (PTEX) has been established as the machinery responsible for the translocation of all classes of exported proteins beyond the parasitophorous vacuolar membrane of the intraerythrocytic malaria parasite. Protein export, particularly in the asexual blood stage, is crucial for parasite survival as exported proteins are involved in remodelling the host cell, an essential process for nutrient uptake, waste removal and immune evasion. Here, we have truncated the conserved C-terminus of one of the essential PTEX components, PTEX150, in Plasmodium falciparum in an attempt to create mutants of reduced functionality. Parasites tolerated C-terminal truncations of up to 125 amino acids with no reduction in growth, protein export or the establishment of new permeability pathways. Quantitative proteomic approaches however revealed a decrease in other PTEX subunits associating with PTEX150 in truncation mutants, suggesting a role for the C-terminus of PTEX150 in regulating PTEX stability. Our analyses also reveal three previously unreported PTEX-associated proteins, namely PV1, Pf113 and Hsp70-x (respective PlasmoDB numbers; PF3D7_1129100, PF3D7_1420700 and PF3D7_0831700) and demonstrate that core PTEX proteins exist in various distinct multimeric forms outside the major complex.


Assuntos
Eritrócitos/parasitologia , Proteínas de Membrana Transportadoras/fisiologia , Plasmodium falciparum/fisiologia , Proteoma/metabolismo , Proteínas de Protozoários/fisiologia , Células Cultivadas , Humanos , Complexos Multiproteicos/metabolismo , Domínios Proteicos , Mapas de Interação de Proteínas , Estabilidade Proteica , Transporte Proteico
19.
J Biol Chem ; 288(38): 27002-27018, 2013 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-23913689

RESUMO

In eukaryotic organisms, cysteine palmitoylation is an important reversible modification that impacts protein targeting, folding, stability, and interactions with partners. Evidence suggests that protein palmitoylation contributes to key biological processes in Apicomplexa with the recent palmitome of the malaria parasite Plasmodium falciparum reporting over 400 substrates that are modified with palmitate by a broad range of protein S-acyl transferases. Dynamic palmitoylation cycles require the action of an acyl-protein thioesterase (APT) that cleaves palmitate from substrates and conveys reversibility to this posttranslational modification. In this work, we identified candidates for APT activity in Toxoplasma gondii. Treatment of parasites with low micromolar concentrations of ß-lactone- or triazole urea-based inhibitors that target human APT1 showed varied detrimental effects at multiple steps of the parasite lytic cycle. The use of an activity-based probe in combination with these inhibitors revealed the existence of several serine hydrolases that are targeted by APT1 inhibitors. The active serine hydrolase, TgASH1, identified as the homologue closest to human APT1 and APT2, was characterized further. Biochemical analysis of TgASH1 indicated that this enzyme cleaves substrates with a specificity similar to APTs, and homology modeling points toward an APT-like enzyme. TgASH1 is dispensable for parasite survival, which indicates that the severe effects observed with the ß-lactone inhibitors are caused by the inhibition of non-TgASH1 targets. Other ASH candidates for APT activity were functionally characterized, and one of them was found to be resistant to gene disruption due to the potential essential nature of the protein.


Assuntos
Inibidores Enzimáticos/farmacologia , Lactonas/farmacologia , Proteínas de Protozoários/antagonistas & inibidores , Tioléster Hidrolases/antagonistas & inibidores , Toxoplasma/enzimologia , Sequência de Aminoácidos , Inibidores Enzimáticos/química , Humanos , Lactonas/química , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Homologia Estrutural de Proteína , Tioléster Hidrolases/química , Tioléster Hidrolases/genética , Tioléster Hidrolases/metabolismo , Toxoplasma/genética , Toxoplasmose/tratamento farmacológico , Toxoplasmose/enzimologia , Toxoplasmose/genética
20.
Nat Commun ; 4: 1415, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23361006

RESUMO

Export of proteins into the infected erythrocyte is critical for malaria parasite survival. The majority of effector proteins are thought to export via a proteinaceous translocon, resident in the parasitophorous vacuole membrane surrounding the parasite. Identification of the Plasmodium translocon of exported proteins and its biochemical association with exported proteins suggests it performs this role. Direct evidence for this, however, is lacking. Here using viable purified Plasmodium falciparum merozoites and three-dimensional structured illumination microscopy, we investigate remodelling events immediately following parasite invasion. We show that multiple complexes of the Plasmodium translocon of exported proteins localize together in foci that dynamically change in clustering behaviour. Furthermore, we provide conclusive evidence of spatial association between exported proteins and exported protein 2, a core component of the Plasmodium translocon of exported proteins, during native conditions and upon generation of translocation intermediates. These data provide the most direct cellular evidence to date that protein export occurs at regions of the parasitophorous vacuole membrane housing the Plasmodium translocon of exported proteins complex.


Assuntos
Eritrócitos/parasitologia , Malária Falciparum/metabolismo , Malária Falciparum/parasitologia , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Animais , Análise por Conglomerados , Eritrócitos/metabolismo , Eritrócitos/patologia , Eritrócitos/ultraestrutura , Proteínas de Fluorescência Verde/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestrutura , Merozoítos/citologia , Merozoítos/metabolismo , Merozoítos/ultraestrutura , Modelos Biológicos , Parasitos/citologia , Parasitos/metabolismo , Parasitos/ultraestrutura , Plasmodium falciparum/citologia , Plasmodium falciparum/ultraestrutura , Estrutura Terciária de Proteína , Transporte Proteico , Desdobramento de Proteína , Proteínas de Protozoários/química , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Vacúolos/metabolismo , Vacúolos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA