Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Biochem ; 84: 813-41, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25621510

RESUMEN

Phylum Apicomplexa comprises a large group of obligate intracellular parasites of high medical and veterinary importance. These organisms succeed intracellularly by effecting remarkable changes in a broad range of diverse host cells. The transformation of the host erythrocyte is particularly striking in the case of the malaria parasite Plasmodium falciparum. P. falciparum exports hundreds of proteins that mediate a complex cellular renovation marked by changes in the permeability, rigidity, and cytoadherence properties of the host erythrocyte. The past decade has seen enormous progress in understanding the identity and function of these exported effectors, as well as the mechanisms by which they are trafficked into the host cell. Here we review these advances, place them in the context of host manipulation by related apicomplexans, and propose key directions for future research.


Asunto(s)
Eritrocitos/parasitología , Plasmodium/fisiología , Animales , Apicomplexa/clasificación , Apicomplexa/fisiología , Humanos , Malaria/inmunología , Malaria/parasitología , Señales de Clasificación de Proteína , Proteínas/metabolismo , Infecciones por Protozoos/inmunología , Infecciones por Protozoos/parasitología , Proteínas Protozoarias/metabolismo
2.
Annu Rev Microbiol ; 76: 67-90, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35417197

RESUMEN

Human malaria, caused by infection with Plasmodium parasites, remains one of the most important global public health problems, with the World Health Organization reporting more than 240 million cases and 600,000 deaths annually as of 2020 (World malaria report 2021). Our understanding of the biology of these parasites is critical for development of effective therapeutics and prophylactics, including both antimalarials and vaccines. Plasmodium is a protozoan organism that is intracellular for most of its life cycle. However, to complete its complex life cycle and to allow for both amplification and transmission, the parasite must egress out of the host cell in a highly regulated manner. This review discusses the major pathways and proteins involved in the egress events during the Plasmodium life cycle-merozoite and gametocyte egress out of red blood cells, sporozoite egress out of the oocyst, and merozoite egress out of the hepatocyte. The similarities, as well as the differences, between the various egress pathways of the parasite highlight both novel cell biology and potential therapeutic targets to arrest its life cycle.


Asunto(s)
Malaria , Parásitos , Plasmodium , Animales , Eritrocitos/metabolismo , Eritrocitos/parasitología , Humanos , Estadios del Ciclo de Vida , Parásitos/metabolismo , Plasmodium/genética , Plasmodium falciparum , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
3.
Cell ; 148(6): 1271-83, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22424234

RESUMEN

Although caused by vastly different pathogens, the world's three most serious infectious diseases, tuberculosis, malaria, and HIV-1 infection, share the common problem of drug resistance. The pace of drug development has been very slow for tuberculosis and malaria and rapid for HIV-1. But for each disease, resistance to most drugs has appeared quickly after the introduction of the drug. Learning how to manage and prevent resistance is a major medical challenge that requires an understanding of the evolutionary dynamics of each pathogen. This Review summarizes the similarities and differences in the evolution of drug resistance for these three pathogens.


Asunto(s)
Resistencia a Medicamentos , Infecciones por VIH/tratamiento farmacológico , Malaria/tratamiento farmacológico , Tuberculosis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , Humanos , Malaria/parasitología , Mycobacterium tuberculosis/efectos de los fármacos , Plasmodium/efectos de los fármacos , Tuberculosis/microbiología
4.
Proc Natl Acad Sci U S A ; 120(26): e2306318120, 2023 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-37307435

RESUMEN

Histidine-rich protein II (HRPII) is secreted by Plasmodium falciparum during the blood stage of malaria infection. High plasma levels of HRPII are associated with cerebral malaria, a severe and highly fatal complication of malaria. HRPII has been shown to induce vascular leakage, the hallmark of cerebral malaria, in blood-brain barrier (BBB) and animal models. We have discovered an important mechanism for BBB disruption that is driven by unique features of HRPII. By characterizing serum from infected patients and HRPII produced by P. falciparum parasites in culture, we found that HRPII exists in large multimeric particles of 14 polypeptides that are richly laden with up to 700 hemes per particle. Heme loading of HRPII is required for efficient binding and internalization via caveolin-mediated endocytosis in hCMEC/D3 cerebral microvascular endothelial cells. Upon acidification of endolysosomes, two-thirds of the hemes are released from acid-labile binding sites and metabolized by heme oxygenase 1, generating ferric iron and reactive oxygen species. Subsequent activation of the NLRP3 inflammasome and IL-1ß secretion resulted in endothelial leakage. Inhibition of these pathways with heme sequestration, iron chelation, or anti-inflammatory drugs protected the integrity of the BBB culture model from HRPII:heme. Increased cerebral vascular permeability was seen after injection of young mice with heme-loaded HRPII (HRPII:heme) but not with heme-depleted HRPII. We propose that during severe malaria infection, HRPII:heme nanoparticles in the bloodstream deliver an overwhelming iron load to endothelial cells to cause vascular inflammation and edema. Disrupting this process is an opportunity for targeted adjunctive therapies to reduce the morbidity and mortality of cerebral malaria.


Asunto(s)
Hemoproteínas , Malaria Cerebral , Malaria Falciparum , Animales , Ratones , Histidina , Células Endoteliales , Inflamación , Hemo , Hierro
5.
J Cell Sci ; 136(6)2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36744402

RESUMEN

N-terminal acetylation is a common eukaryotic protein modification that involves the addition of an acetyl group to the N-terminus of a polypeptide. This modification is largely performed by cytosolic N-terminal acetyltransferases (NATs). Most associate with the ribosome, acetylating nascent polypeptides co-translationally. In the malaria parasite Plasmodium falciparum, exported effectors are thought to be translated into the endoplasmic reticulum (ER), processed by the aspartic protease plasmepsin V and then N-acetylated, despite having no clear access to cytosolic NATs. Here, we used inducible gene deletion and post-transcriptional knockdown to investigate the primary ER-resident NAT candidate, Pf3D7_1437000. We found that it localizes to the ER and is required for parasite growth. However, depletion of Pf3D7_1437000 had no effect on protein export or acetylation of the exported proteins HRP2 and HRP3. Despite this, Pf3D7_1437000 depletion impedes parasite development within the host red blood cell and prevents parasites from completing genome replication. Thus, this work provides further proof of N-terminal acetylation of secretory system proteins, a process unique to apicomplexan parasites, but strongly discounts a promising candidate for this post-translational modification.


Asunto(s)
Acetiltransferasas , Retículo Endoplásmico , Plasmodium falciparum , Acetiltransferasas/metabolismo , Retículo Endoplásmico/metabolismo , Péptidos/metabolismo , Plasmodium falciparum/enzimología , Procesamiento Proteico-Postraduccional , Proteínas Protozoarias/metabolismo
6.
Nature ; 561(7721): 70-75, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30150771

RESUMEN

The putative Plasmodium translocon of exported proteins (PTEX) is essential for transport of malarial effector proteins across a parasite-encasing vacuolar membrane into host erythrocytes, but the mechanism of this process remains unknown. Here we show that PTEX is a bona fide translocon by determining structures of the PTEX core complex at near-atomic resolution using cryo-electron microscopy. We isolated the endogenous PTEX core complex containing EXP2, PTEX150 and HSP101 from Plasmodium falciparum in the 'engaged' and 'resetting' states of endogenous cargo translocation using epitope tags inserted using the CRISPR-Cas9 system. In the structures, EXP2 and PTEX150 interdigitate to form a static, funnel-shaped pseudo-seven-fold-symmetric protein-conducting channel spanning the vacuolar membrane. The spiral-shaped AAA+ HSP101 hexamer is tethered above this funnel, and undergoes pronounced compaction that allows three of six tyrosine-bearing pore loops lining the HSP101 channel to dissociate from the cargo, resetting the translocon for the next threading cycle. Our work reveals the mechanism of P. falciparum effector export, and will inform structure-based design of drugs targeting this unique translocon.


Asunto(s)
Microscopía por Crioelectrón , Plasmodium falciparum/ultraestructura , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/ultraestructura , Animales , Eritrocitos/metabolismo , Eritrocitos/parasitología , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Modelos Biológicos , Modelos Moleculares , Terapia Molecular Dirigida/tendencias , Movimiento , Plasmodium falciparum/química , Plasmodium falciparum/metabolismo , Unión Proteica , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Transporte de Proteínas , Proteínas Protozoarias/química , Vacuolas/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34446549

RESUMEN

The RhopH complex is implicated in malaria parasites' ability to invade and create new permeability pathways in host erythrocytes, but its mechanisms remain poorly understood. Here, we enrich the endogenous RhopH complex in a native soluble form, comprising RhopH2, CLAG3.1, and RhopH3, directly from parasite cell lysates and determine its atomic structure using cryo-electron microscopy (cryo-EM), mass spectrometry, and the cryoID program. CLAG3.1 is positioned between RhopH2 and RhopH3, which both share substantial binding interfaces with CLAG3.1 but make minimal contacts with each other. The forces stabilizing individual subunits include 13 intramolecular disulfide bonds. Notably, CLAG3.1 residues 1210 to 1223, previously predicted to constitute a transmembrane helix, are embedded within a helical bundle formed by residues 979 to 1289 near the C terminus of CLAG3.1. Buried in the core of the RhopH complex and largely shielded from solvent, insertion of this putative transmembrane helix into the erythrocyte membrane would likely require a large conformational rearrangement. Given the unusually high disulfide content of the complex, it is possible that such a rearrangement could be initiated by the breakage of allosteric disulfide bonds, potentially triggered by interactions at the erythrocyte membrane. This first direct observation of an exported Plasmodium falciparum transmembrane protein-in a soluble, trafficking state and with atomic details of buried putative membrane-insertion helices-offers insights into the assembly and trafficking of RhopH and other parasite-derived complexes to the erythrocyte membrane. Our study demonstrates the potential the endogenous structural proteomics approach holds for elucidating the molecular mechanisms of hard-to-isolate complexes in their native, functional forms.


Asunto(s)
Membrana Eritrocítica/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/química , Permeabilidad de la Membrana Celular , Microscopía por Crioelectrón , Membrana Eritrocítica/parasitología , Humanos , Modelos Moleculares , Nutrientes/metabolismo , Conformación Proteica , Proteómica , Proteínas Protozoarias/fisiología , Proteínas Protozoarias/ultraestructura , Relación Estructura-Actividad
8.
J Biol Chem ; 298(9): 102355, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35952758

RESUMEN

Plasmepsin V (PM V) is a pepsin-like aspartic protease essential for growth of the malarial parasite Plasmodium falciparum. Previous work has shown PM V to be an endoplasmic reticulum-resident protease that processes parasite proteins destined for export into the host cell. Depletion or inhibition of the enzyme is lethal during asexual replication within red blood cells as well as during the formation of sexual stage gametocytes. The structure of the Plasmodium vivax PM V has been characterized by X-ray crystallography, revealing a canonical pepsin fold punctuated by structural features uncommon to secretory aspartic proteases; however, the function of this unique structure is unclear. Here, we used parasite genetics to probe these structural features by attempting to rescue lethal PM V depletion with various mutant enzymes. We found an unusual nepenthesin 1-type insert in the PM V gene to be essential for parasite growth and PM V activity. Mutagenesis of the nepenthesin insert suggests that both its amino acid sequence and one of the two disulfide bonds that undergird its structure are required for the insert's role in PM V function. Furthermore, molecular dynamics simulations paired with Markov state modeling suggest that mutations to the nepenthesin insert may allosterically affect PM V catalysis through multiple mechanisms. Taken together, these data provide further insights into the structure of the P. falciparum PM V protease.


Asunto(s)
Malaria Falciparum , Plasmodium falciparum , Ácido Aspártico Endopeptidasas/metabolismo , Disulfuros/metabolismo , Humanos , Pepsina A/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo
9.
J Biol Chem ; 298(2): 101550, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973333

RESUMEN

The malaria-causing parasite Plasmodium falciparum is responsible for over 200 million infections and 400,000 deaths per year. At multiple stages during its complex life cycle, P. falciparum expresses several essential proteins tethered to its surface by glycosylphosphatidylinositol (GPI) anchors, which are critical for biological processes such as parasite egress and reinvasion of host red blood cells. Targeting this pathway therapeutically has the potential to broadly impact parasite development across several life stages. Here, we characterize an upstream component of parasite GPI anchor biosynthesis, the putative phosphomannomutase (PMM) (EC 5.4.2.8), HAD5 (PF3D7_1017400). We confirmed the PMM and phosphoglucomutase activities of purified recombinant HAD5 by developing novel linked enzyme biochemical assays. By regulating the expression of HAD5 in transgenic parasites with a TetR-DOZI-inducible knockdown system, we demonstrated that HAD5 is required for malaria parasite egress and erythrocyte reinvasion, and we assessed the role of HAD5 in GPI anchor synthesis by autoradiography of radiolabeled glucosamine and thin layer chromatography. Finally, we determined the three-dimensional X-ray crystal structure of HAD5 and identified a substrate analog that specifically inhibits HAD5 compared to orthologous human PMMs in a time-dependent manner. These findings demonstrate that the GPI anchor biosynthesis pathway is exceptionally sensitive to inhibition in parasites and that HAD5 has potential as a specific, multistage antimalarial target.


Asunto(s)
Fosfotransferasas (Fosfomutasas) , Plasmodium falciparum , Proteínas Protozoarias , Animales , Eritrocitos/parasitología , Glicosilfosfatidilinositoles/metabolismo , Humanos , Malaria Falciparum/parasitología , Fosfotransferasas (Fosfomutasas)/genética , Fosfotransferasas (Fosfomutasas)/metabolismo , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
10.
Nat Methods ; 17(1): 79-85, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31768063

RESUMEN

X-ray crystallography often requires non-native constructs involving mutations or truncations, and is challenged by membrane proteins and large multicomponent complexes. We present here a bottom-up endogenous structural proteomics approach whereby near-atomic-resolution cryo electron microscopy (cryoEM) maps are reconstructed ab initio from unidentified protein complexes enriched directly from the endogenous cellular milieu, followed by identification and atomic modeling of the proteins. The proteins in each complex are identified using cryoID, a program we developed to identify proteins in ab initio cryoEM maps. As a proof of principle, we applied this approach to the malaria-causing parasite Plasmodium falciparum, an organism that has resisted conventional structural-biology approaches, to obtain atomic models of multiple protein complexes implicated in intraerythrocytic survival of the parasite. Our approach is broadly applicable for determining structures of undiscovered protein complexes enriched directly from endogenous sources.


Asunto(s)
Microscopía por Crioelectrón/métodos , Eritrocitos/parasitología , Procesamiento de Imagen Asistido por Computador/métodos , Complejos Multiproteicos/química , Plasmodium falciparum/metabolismo , Proteómica/métodos , Proteínas Protozoarias/química , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Humanos , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Espectrometría de Masas , Modelos Moleculares , Complejos Multiproteicos/ultraestructura , Plasmodium falciparum/aislamiento & purificación , Conformación Proteica , Proteínas Protozoarias/ultraestructura
11.
J Biol Chem ; 295(25): 8425-8441, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32366462

RESUMEN

Plasmepsins are a group of diverse aspartic proteases in the malaria parasite Plasmodium Their functions are strikingly multifaceted, ranging from hemoglobin degradation to secretory organelle protein processing for egress, invasion, and effector export. Some, particularly the digestive vacuole plasmepsins, have been extensively characterized, whereas others, such as the transmission-stage plasmepsins, are minimally understood. Some (e.g. plasmepsin V) have exquisite cleavage sequence specificity; others are fairly promiscuous. Some have canonical pepsin-like aspartic protease features, whereas others have unusual attributes, including the nepenthesin loop of plasmepsin V and a histidine in place of a catalytic aspartate in plasmepsin III. We have learned much about the functioning of these enzymes, but more remains to be discovered about their cellular roles and even their mechanisms of action. Their importance in many key aspects of parasite biology makes them intriguing targets for antimalarial chemotherapy. Further consideration of their characteristics suggests that some are more viable drug targets than others. Indeed, inhibitors of invasion and egress offer hope for a desperately needed new drug to combat this nefarious organism.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Plasmodium/metabolismo , Proteínas Protozoarias/metabolismo , Antimaláricos/química , Antimaláricos/metabolismo , Antimaláricos/uso terapéutico , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/química , Proteasas de Ácido Aspártico/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Humanos , Malaria/tratamiento farmacológico , Malaria/parasitología , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/química
12.
Cell Microbiol ; 22(5): e13168, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31990132

RESUMEN

Intraerythrocytic malaria parasites reside within a parasitophorous vacuole membrane (PVM) that closely overlays the parasite plasma membrane. Although the PVM is the site of several transport activities essential to parasite survival, the basis for organisation of this membrane system is unknown. Here, we performed proximity labeling at the PVM with BioID2, which highlighted a group of single-pass integral membrane proteins that constitute a major component of the PVM proteome but whose function remains unclear. We investigated EXP1, the longest known member of this group, by adapting a CRISPR/Cpf1 genome editing system to install the TetR-DOZI-aptamers system for conditional translational control. Importantly, although EXP1 was required for intraerythrocytic development, a previously reported in vitro glutathione S-transferase activity could not account for this essential EXP1 function in vivo. EXP1 knockdown was accompanied by profound changes in vacuole ultrastructure, including apparent increased separation of the PVM from the parasite plasma membrane and formation of abnormal membrane structures. Furthermore, although activity of the Plasmodium translocon of exported proteins was not impacted by depletion of EXP1, the distribution of the translocon pore-forming protein EXP2 but not the HSP101 unfoldase was substantially altered. Collectively, our results reveal a novel PVM defect that indicates a critical role for EXP1 in maintaining proper organisation of EXP2 within the PVM.


Asunto(s)
Antígenos de Protozoos/inmunología , Parásitos/genética , Parásitos/metabolismo , Vacuolas/parasitología , Animales , Antígenos de Protozoos/genética , Edición Génica , Malaria/parasitología , Proteínas de la Membrana/metabolismo , Plasmodium/genética , Plasmodium/metabolismo , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Vacuolas/metabolismo
13.
PLoS Pathog ; 14(5): e1007031, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29768491

RESUMEN

Parasite egress from infected erythrocytes and invasion of new red blood cells are essential processes for the exponential asexual replication of the malaria parasite. These two tightly coordinated events take place in less than a minute and are in part regulated and mediated by proteases. Dipeptidyl aminopeptidases (DPAPs) are papain-fold cysteine proteases that cleave dipeptides from the N-terminus of protein substrates. DPAP3 was previously suggested to play an essential role in parasite egress. However, little is known about its enzymatic activity, intracellular localization, or biological function. In this study, we recombinantly expressed DPAP3 and demonstrate that it has indeed dipeptidyl aminopeptidase activity, but contrary to previously studied DPAPs, removal of its internal prodomain is not required for activation. By combining super resolution microscopy, time-lapse fluorescence microscopy, and immunoelectron microscopy, we show that Plasmodium falciparum DPAP3 localizes to apical organelles that are closely associated with the neck of the rhoptries, and from which DPAP3 is secreted immediately before parasite egress. Using a conditional knockout approach coupled to complementation studies with wild type or mutant DPAP3, we show that DPAP3 activity is important for parasite proliferation and critical for efficient red blood cell invasion. We also demonstrate that DPAP3 does not play a role in parasite egress, and that the block in egress phenotype previously reported for DPAP3 inhibitors is due to off target or toxicity effects. Finally, using a flow cytometry assay to differentiate intracellular parasites from extracellular parasites attached to the erythrocyte surface, we show that DPAP3 is involved in the initial attachment of parasites to the red blood cell surface. Overall, this study establishes the presence of a DPAP3-dependent invasion pathway in malaria parasites.


Asunto(s)
Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Malaria Falciparum/parasitología , Plasmodium falciparum/enzimología , Animales , Proteasas de Cisteína/metabolismo , Eritrocitos/microbiología , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Malaria Falciparum/metabolismo , Malaria Falciparum/patología , Merozoítos/metabolismo , Merozoítos/fisiología , Orgánulos/metabolismo , Péptido Hidrolasas/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Proteolisis , Proteínas Protozoarias/metabolismo
14.
Nature ; 511(7511): 592-5, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25043010

RESUMEN

To mediate its survival and virulence, the malaria parasite Plasmodium falciparum exports hundreds of proteins into the host erythrocyte. To enter the host cell, exported proteins must cross the parasitophorous vacuolar membrane (PVM) within which the parasite resides, but the mechanism remains unclear. A putative Plasmodium translocon of exported proteins (PTEX) has been suggested to be involved for at least one class of exported proteins; however, direct functional evidence for this has been elusive. Here we show that export across the PVM requires heat shock protein 101 (HSP101), a ClpB-like AAA+ ATPase component of PTEX. Using a chaperone auto-inhibition strategy, we achieved rapid, reversible ablation of HSP101 function, resulting in a nearly complete block in export with substrates accumulating in the vacuole in both asexual and sexual parasites. Surprisingly, this block extended to all classes of exported proteins, revealing HSP101-dependent translocation across the PVM as a convergent step in the multi-pathway export process. Under export-blocked conditions, association between HSP101 and other components of the PTEX complex was lost, indicating that the integrity of the complex is required for efficient protein export. Our results demonstrate an essential and universal role for HSP101 in protein export and provide strong evidence for PTEX function in protein translocation into the host cell.


Asunto(s)
Eritrocitos/parasitología , Proteínas de Choque Térmico/metabolismo , Interacciones Huésped-Parásitos/fisiología , Malaria Falciparum/parasitología , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Estadios del Ciclo de Vida/genética , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Transporte de Proteínas , Vacuolas/parasitología
15.
Annu Rev Microbiol ; 68: 259-78, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25002093

RESUMEN

For over a century, heme metabolism has been recognized to play a central role during intraerythrocytic infection by Plasmodium parasites, the causative agent of malaria. Parasites liberate vast quantities of potentially cytotoxic heme as a by-product of hemoglobin catabolism within the digestive vacuole, where heme is predominantly sequestered as inert crystalline hemozoin. Plasmodium spp. also utilize heme as a metabolic cofactor. Despite access to abundant host-derived heme, parasites paradoxically maintain a biosynthetic pathway. This pathway has been assumed to produce the heme incorporated into mitochondrial cytochromes that support electron transport. In this review, we assess our current understanding of the love-hate relationship between Plasmodium parasites and heme, we discuss recent studies that clarify several long-standing riddles about heme production and utilization by parasites, and we consider remaining challenges and opportunities for understanding and targeting heme metabolism within parasites.


Asunto(s)
Hemo/metabolismo , Malaria/parasitología , Plasmodium/metabolismo , Animales , Humanos , Malaria/metabolismo
16.
Cell Microbiol ; 20(10): e12868, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29900649

RESUMEN

Because Plasmodium falciparum replicates inside of a parasitophorous vacuole (PV) within a human erythrocyte, parasite egress requires the rupture of two limiting membranes. Parasite Ca2+ , kinases, and proteases contribute to efficient egress; their coordination in space and time is not known. Here, the kinetics of parasite egress were linked to specific steps with specific compartment markers, using live-cell microscopy of parasites expressing PV-targeted fluorescent proteins, and specific egress inhibitors. Several minutes before egress, under control of parasite [Ca2+ ]i , the PV began rounding. Then after ~1.5 min, under control of PfPKG and SUB1, there was abrupt rupture of the PV membrane and release of vacuolar contents. Over the next ~6 min, there was progressive vacuolar membrane deterioration simultaneous with erythrocyte membrane distortion, lasting until the final minute of the egress programme when newly formed parasites mobilised and erythrocyte membranes permeabilised and then ruptured-a dramatic finale to the parasite cycle of replication.


Asunto(s)
Membrana Eritrocítica/parasitología , Eritrocitos/patología , Eritrocitos/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Vacuolas/parasitología , Calcio/metabolismo , Colorantes Fluorescentes , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/patología , Plasmodium falciparum/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Vacuolas/metabolismo
17.
Malar J ; 18(1): 78, 2019 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-30866956

RESUMEN

BACKGROUND: The Plasmodium falciparum histidine-rich protein II (PfHRP2) is a common biomarker used in malaria rapid diagnostic tests (RDTs), but can persist in the blood for up to 40 days following curative treatment. The persistence of PfHRP2 presents a false positive limitation to diagnostic interpretation. However, the in vivo dynamics and compartmentalization underlying PfHRP2 persistence have not been fully characterized in the plasma and erythrocyte (RBC) fraction of the whole blood. METHODS: The kinetics and persistence of PfHRP2 in the plasma and RBC fractions of the whole blood were investigated post-treatment in human clinical samples and samples isolated from BALB/c mice infected with a novel transgenic Plasmodium berghei parasite engineered to express PfHRP2 (PbPfHRP2). RESULTS: PfHRP2 levels in human RBCs were consistently 20-40 times greater than plasma levels, even post-parasite clearance. PfHRP2 positive, DNA negative, once-infected RBCs were identified in patients that comprised 0.1-1% of total RBCs for 6 and 12 days post-treatment, even post-atovaquone-proguanil regimens. Transgenic PbPfHRP2 parasites in BALB/c mice produced and exported tgPfHRP2 to the RBC cytosol similar to P. falciparum. As in humans, tgPfHRP2 levels were found to be approximately 20-fold higher within the RBC fraction than the plasma post-treatment. RBC localized tgPfHRP2 persisted longer than tgPfHRP2 in the plasma after curative treatment. tgPfHRP2 positive, but DNA negative once-infected RBCs were also detected in mouse peripheral blood for 7-9 days after curative treatment. CONCLUSIONS: The data suggest that persistence of PfHRP2 is due to slower clearance of protein from the RBC fraction of the whole blood. This appears to be a result of the presence PfHRP2 in previously infected, pitted cells, as opposed to PfHRP2 binding naïve RBCs in circulation post-treatment. The results thus confirm that the extended duration of RDT positivity after parasite clearance is likely due to pitted, once-infected RBCs that remain positive for PfHRP2.


Asunto(s)
Antígenos de Protozoos/sangre , Antimaláricos/administración & dosificación , Eritrocitos/química , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/patología , Plasma/química , Proteínas Protozoarias/sangre , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo
18.
Cell Microbiol ; 19(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28067475

RESUMEN

The malaria parasite exports numerous proteins into its host red blood cell (RBC). The trafficking of these exported effectors is complex. Proteins are first routed through the secretory system, into the parasitophorous vacuole (PV), a membranous compartment enclosing the parasite. Proteins are then translocated across the PV membrane in a process requiring ATP and unfolding. Once in the RBC compartment the exported proteins are then refolded and further trafficked to their final localizations. Chaperones are important in the unfolding and refolding processes. Recently, it was suggested that the parasite TRiC chaperonin complex is exported, and that it is involved in trafficking of exported effectors. Using a parasite-specific antibody and epitope-tagged transgenic parasites we could observe no export of Plasmodium TRiC into the RBC. We tested the importance of the parasite TRiC by creating a regulatable knockdown line of the TRiC-θ subunit. Loss of the parasite TRiC-θ led to a severe growth defect in asexual development, but did not alter protein export into the RBC. These observations indicate that the TRiC proteins play a critical role in parasite biology, though their function, within the parasite, appears unrelated to protein trafficking in the RBC compartment.


Asunto(s)
Chaperoninas/metabolismo , Citosol/metabolismo , Malaria Falciparum/patología , Complejos Multiproteicos/metabolismo , Plasmodium falciparum/patogenicidad , Membrana Celular/metabolismo , Eritrocitos/parasitología , Regulación de la Expresión Génica/genética , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Replegamiento Proteico , Transporte de Proteínas/fisiología , Vacuolas/parasitología
19.
Bioorg Med Chem Lett ; 28(10): 1846-1848, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29691139

RESUMEN

The human parasite Plasmodium falciparum kills an estimated 445,000 people a year, with the most fatalities occurring in African children. Previous studies identified falcilysin (FLN) as a malarial metalloprotease essential for parasite development in the human host. Despite its essentiality, the biological roles of this protease are not well understood. Here we describe the optimization of a piperazine-based hydroxamic acid scaffold to develop the first reported inhibitors of FLN. Inhibitors were tested against cultured parasites, and parasiticidal activity correlated with potency against FLN. This suggests these compounds kill P. falciparum by blocking FLN, and that FLN is a druggable target. These compounds represent an important step towards validating FLN as a therapeutic target and towards the development of chemical tools to investigate the function of this protease.


Asunto(s)
Antimaláricos/química , Ácidos Hidroxámicos/química , Metaloendopeptidasas/antagonistas & inhibidores , Piperazina/química , Inhibidores de Proteasas/química , Proteínas Protozoarias/antagonistas & inhibidores , Antimaláricos/síntesis química , Antimaláricos/farmacología , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/farmacología , Concentración 50 Inhibidora , Metaloendopeptidasas/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Inhibidores de Proteasas/síntesis química , Inhibidores de Proteasas/farmacología , Proteínas Protozoarias/metabolismo , Relación Estructura-Actividad
20.
Biochemistry ; 55(34): 4836-49, 2016 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-27490825

RESUMEN

Heme oxygenase (HO) is a ubiquitous enzyme with key roles in inflammation, cell signaling, heme disposal, and iron acquisition. HO catalyzes the oxidative conversion of heme to biliverdin (BV) using a conserved histidine to coordinate the iron atom of bound heme. This His-heme interaction has been regarded as being essential for enzyme activity, because His-to-Ala mutants fail to convert heme to biliverdin in vitro. We probed a panel of proximal His mutants of cyanobacterial, human, and plant HO enzymes using a live-cell activity assay based on heterologous co-expression in Escherichia coli of each HO mutant and a fluorescent biliverdin biosensor. In contrast to in vitro studies with purified proteins, we observed that multiple HO mutants retained significant activity within the intracellular environment of bacteria. X-ray crystallographic structures of human HO1 H25R with bound heme and additional functional studies suggest that HO mutant activity inside these cells does not involve heme ligation by a proximal amino acid. Our study reveals unexpected plasticity in the active site binding interactions with heme that can support HO activity within cells, suggests important contributions by the surrounding active site environment to HO catalysis, and can guide efforts to understand the evolution and divergence of HO function.


Asunto(s)
Hemo Oxigenasa (Desciclizante)/química , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/química , Hemo-Oxigenasa 1/metabolismo , Sustitución de Aminoácidos , Arabidopsis/enzimología , Arabidopsis/genética , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biliverdina/metabolismo , Biocatálisis , Catalasa/metabolismo , Dominio Catalítico/genética , Cristalografía por Rayos X , Escherichia coli/enzimología , Escherichia coli/genética , Hemo/metabolismo , Hemo Oxigenasa (Desciclizante)/genética , Hemo-Oxigenasa 1/genética , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Conformación Proteica , Synechocystis/enzimología , Synechocystis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA