Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(28): e2403442121, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38968107

RESUMEN

Plasmodium falciparum causes severe malaria and assembles a protein translocon (PTEX) complex at the parasitophorous vacuole membrane (PVM) of infected erythrocytes, through which several hundred proteins are exported to facilitate growth. The preceding liver stage of infection involves growth in a hepatocyte-derived PVM; however, the importance of protein export during P. falciparum liver infection remains unexplored. Here, we use the FlpL/FRT system to conditionally excise genes in P. falciparum sporozoites for functional liver-stage studies. Disruption of PTEX members ptex150 and exp2 did not affect sporozoite development in mosquitoes or infectivity for hepatocytes but attenuated liver-stage growth in humanized mice. While PTEX150 deficiency reduced fitness on day 6 postinfection by 40%, EXP2 deficiency caused 100% loss of liver parasites, demonstrating that PTEX components are required for growth in hepatocytes to differing degrees. To characterize PTEX loss-of-function mutations, we localized four liver-stage Plasmodium export element (PEXEL) proteins. P. falciparum liver specific protein 2 (LISP2), liver-stage antigen 3 (LSA3), circumsporozoite protein (CSP), and a Plasmodium berghei LISP2 reporter all localized to the periphery of P. falciparum liver stages but were not exported beyond the PVM. Expression of LISP2 and CSP but not LSA3 was reduced in ptex150-FRT and exp2-FRT liver stages, suggesting that expression of some PEXEL proteins is affected directly or indirectly by PTEX disruption. These results show that PTEX150 and EXP2 are important for P. falciparum development in hepatocytes and emphasize the emerging complexity of PEXEL protein trafficking.


Asunto(s)
Hepatocitos , Hígado , Malaria Falciparum , Plasmodium falciparum , Proteínas Protozoarias , Esporozoítos , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Animales , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Esporozoítos/metabolismo , Esporozoítos/crecimiento & desarrollo , Ratones , Hígado/parasitología , Hígado/metabolismo , Humanos , Hepatocitos/parasitología , Hepatocitos/metabolismo , Malaria Falciparum/parasitología
2.
Mol Microbiol ; 121(4): 717-726, 2024 04.
Artículo en Inglés | MEDLINE | ID: mdl-38225194

RESUMEN

Apicomplexan parasites are aetiological agents of numerous diseases in humans and livestock. Functional genomics studies in these parasites enable the identification of biological mechanisms and protein functions that can be targeted for therapeutic intervention. Recent improvements in forward genetics and whole-genome screens utilising CRISPR/Cas technology have revolutionised the functional analysis of genes during Apicomplexan infection of host cells. Here, we highlight key discoveries from CRISPR/Cas9 screens in Apicomplexa or their infected host cells and discuss remaining challenges to maximise this technology that may help answer fundamental questions about parasite-host interactions.


Asunto(s)
Apicomplexa , Parásitos , Humanos , Animales , Sistemas CRISPR-Cas , Genoma , Apicomplexa/genética , Parásitos/genética , Interacciones Huésped-Parásitos
3.
Gastroenterology ; 163(6): 1643-1657.e14, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36037995

RESUMEN

BACKGROUND & AIMS: Necroptosis is a highly inflammatory mode of cell death that has been implicated in causing hepatic injury including steatohepatitis/ nonalcoholic steatohepatitis (NASH); however, the evidence supporting these claims has been controversial. A comprehensive, fundamental understanding of cell death pathways involved in liver disease critically underpins rational strategies for therapeutic intervention. We sought to define the role and relevance of necroptosis in liver pathology. METHODS: Several animal models of human liver pathology, including diet-induced steatohepatitis in male mice and diverse infections in both male and female mice, were used to dissect the relevance of necroptosis in liver pathobiology. We applied necroptotic stimuli to primary mouse and human hepatocytes to measure their susceptibility to necroptosis. Paired liver biospecimens from patients with NASH, before and after intervention, were analyzed. DNA methylation sequencing was also performed to investigate the epigenetic regulation of RIPK3 expression in primary human and mouse hepatocytes. RESULTS: Identical infection kinetics and pathologic outcomes were observed in mice deficient in an essential necroptotic effector protein, MLKL, compared with control animals. Mice lacking MLKL were indistinguishable from wild-type mice when fed a high-fat diet to induce NASH. Under all conditions tested, we were unable to induce necroptosis in hepatocytes. We confirmed that a critical activator of necroptosis, RIPK3, was epigenetically silenced in mouse and human primary hepatocytes and rendered them unable to undergo necroptosis. CONCLUSIONS: We have provided compelling evidence that necroptosis is disabled in hepatocytes during homeostasis and in the pathologic conditions tested in this study.


Asunto(s)
Necroptosis , Enfermedad del Hígado Graso no Alcohólico , Humanos , Femenino , Masculino , Ratones , Animales , Epigénesis Genética , Enfermedad del Hígado Graso no Alcohólico/genética , Hepatocitos , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Proteínas Quinasas/genética
4.
Cell Microbiol ; 23(3): e13289, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33197142

RESUMEN

Twenty years ago the Molecular Approaches to Malaria conference was conceived as a forum to present the very latest advances in malaria research and to consolidate and forge new collaborative links between international researchers. The 6th MAM conference, held in February 2020 in Australia, provided 5 days of stimulating scientific exchange and highlighted the incredible malaria research conducted globally that is providing the critical knowledge and cutting-edge technological tools needed to control and ultimately eliminate malaria.


Asunto(s)
Malaria , Plasmodium , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Desarrollo de Medicamentos , Resistencia a Medicamentos , Humanos , Inmunogenicidad Vacunal , Malaria/tratamiento farmacológico , Malaria/inmunología , Malaria/parasitología , Malaria/prevención & control , Vacunas contra la Malaria/inmunología , Plasmodium/efectos de los fármacos , Plasmodium/genética , Plasmodium/patogenicidad , Plasmodium/fisiología
5.
Bioorg Chem ; 117: 105359, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34689083

RESUMEN

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.


Asunto(s)
Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Antimaláricos/química , Antimaláricos/farmacología , Malaria/tratamiento farmacológico , Plasmodium/efectos de los fármacos , Quinazolinas/química , Quinazolinas/farmacología , Aminación , Compuestos de Anilina/uso terapéutico , Animales , Antimaláricos/uso terapéutico , Femenino , Humanos , Malaria/parasitología , Ratones , Plasmodium/fisiología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Quinazolinas/uso terapéutico
6.
Mol Microbiol ; 109(4): 458-473, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29873127

RESUMEN

Transmission of the malaria parasite Plasmodium falciparum involves infection of Anopheles mosquitoes. Here we characterize SOPT, a protein expressed in P. falciparum ookinetes that facilitates infection of the mosquito midgut. SOPT was identified on the basis that it contains a signal peptide, a PEXEL-like sequence and is expressed in asexual, ookinete and sporozoite stages, suggesting it is involved in infecting the human or mosquito host. SOPT is predicted to contain a subtilisin-like fold with a non-canonical catalytic triad and is orthologous to P. berghei PIMMS2. Localization studies reveal that SOPT is not exported to the erythrocyte but is expressed in ookinetes at the parasite periphery. SOPT-deficient parasites develop normally through the asexual and sexual stages and produce equivalent numbers of ookinetes to NF54 controls, however, they form fewer oocysts and sporozoites in mosquitoes. SOPT-deficient parasites were also unable to activate the immune-responsive midgut invasion marker SRPN6 after mosquito ingestion, suggesting they are defective for entry into the midgut. Disruption of SOPT in P. berghei (PIMMS2) did not affect other lifecycle stages or ookinete development but again resulted in fewer oocysts and sporozoites in mosquitoes. Collectively, this study shows that SOPT/PIMMS2 plays a conserved role in ookinetes of different Plasmodium species.


Asunto(s)
Anopheles/parasitología , Sistema Digestivo/parasitología , Oocistos/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Esporozoítos/crecimiento & desarrollo , Animales , Malaria Falciparum/transmisión , Mosquitos Vectores/parasitología , Subtilisina/metabolismo
7.
Genome Res ; 25(1): 129-41, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25236617

RESUMEN

Burkholderia pseudomallei (Bp) is the causative agent of the infectious disease melioidosis. To investigate population diversity, recombination, and horizontal gene transfer in closely related Bp isolates, we performed whole-genome sequencing (WGS) on 106 clinical, animal, and environmental strains from a restricted Asian locale. Whole-genome phylogenies resolved multiple genomic clades of Bp, largely congruent with multilocus sequence typing (MLST). We discovered widespread recombination in the Bp core genome, involving hundreds of regions associated with multiple haplotypes. Highly recombinant regions exhibited functional enrichments that may contribute to virulence. We observed clade-specific patterns of recombination and accessory gene exchange, and provide evidence that this is likely due to ongoing recombination between clade members. Reciprocally, interclade exchanges were rarely observed, suggesting mechanisms restricting gene flow between clades. Interrogation of accessory elements revealed that each clade harbored a distinct complement of restriction-modification (RM) systems, predicted to cause clade-specific patterns of DNA methylation. Using methylome sequencing, we confirmed that representative strains from separate clades indeed exhibit distinct methylation profiles. Finally, using an E. coli system, we demonstrate that Bp RM systems can inhibit uptake of non-self DNA. Our data suggest that RM systems borne on mobile elements, besides preventing foreign DNA invasion, may also contribute to limiting exchanges of genetic material between individuals of the same species. Genomic clades may thus represent functional units of genetic isolation in Bp, modulating intraspecies genetic diversity.


Asunto(s)
Burkholderia pseudomallei/genética , Epigénesis Genética , Genoma Bacteriano , Recombinación Genética , Transcriptoma , Animales , Cartilla de ADN , ADN Bacteriano/genética , Escherichia coli/genética , Femenino , Eliminación de Gen , Estudios de Asociación Genética , Genómica , Haplotipos , Humanos , Melioidosis/microbiología , Ratones , Ratones Endogámicos BALB C , Tipificación de Secuencias Multilocus , Filogenia , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
8.
Annu Rev Microbiol ; 67: 243-69, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23808341

RESUMEN

One of the most fascinating and remarkable features of Plasmodium parasites, which cause malaria, is their choice of erythrocytes as the principal host cells in which to reside during infection of a vertebrate host. Parasites completely renovate the terminally differentiated cells, which lack most of the normal organelles and functions of other cells, such as a nucleus and the machinery to express and transport proteins to subcellular locations. Erythrocyte remodeling begins immediately after invasion by the Plasmodium parasite, by expression and export of many hundreds of proteins that assemble into molecular machinery in the host cell that permit protein trafficking, harvesting of nutrients, and mechanisms to evade host immune responses. In this review, we discuss recent studies on erythrocyte remodeling, including mechanisms of protein export as well as the identity, functions, and subcellular locations of key exported proteins.


Asunto(s)
Eritrocitos/parasitología , Malaria/parasitología , Plasmodium/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Humanos , Plasmodium/genética , Transporte de Proteínas , Proteínas Protozoarias/genética
9.
Cell Microbiol ; 19(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28371168

RESUMEN

The malaria sporozoite injected by a mosquito migrates to the liver by traversing host cells. The sporozoite also traverses hepatocytes before invading a terminal hepatocyte and developing into exoerythrocytic forms. Hepatocyte infection is critical for parasite development into merozoites that infect erythrocytes, and the sporozoite is thus an important target for antimalarial intervention. Here, we investigated two abundant sporozoite proteins of the most virulent malaria parasite Plasmodium falciparum and show that they play important roles during cell traversal and invasion of human hepatocytes. Incubation of P. falciparum sporozoites with R1 peptide, an inhibitor of apical merozoite antigen 1 (AMA1) that blocks merozoite invasion of erythrocytes, strongly reduced cell traversal activity. Consistent with its inhibitory effect on merozoites, R1 peptide also reduced sporozoite entry into human hepatocytes. The strong but incomplete inhibition prompted us to study the AMA-like protein, merozoite apical erythrocyte-binding ligand (MAEBL). MAEBL-deficient P. falciparum sporozoites were severely attenuated for cell traversal activity and hepatocyte entry in vitro and for liver infection in humanized chimeric liver mice. This study shows that AMA1 and MAEBL are important for P. falciparum sporozoites to perform typical functions necessary for infection of human hepatocytes. These two proteins therefore have important roles during infection at distinct points in the life cycle, including the blood, mosquito, and liver stages.


Asunto(s)
Hepatocitos/parasitología , Malaria Falciparum/parasitología , Proteínas de la Membrana/antagonistas & inhibidores , Merozoítos/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/antagonistas & inhibidores , Receptores de Superficie Celular/antagonistas & inhibidores , Esporozoítos/crecimiento & desarrollo , Animales , Anopheles/parasitología , Antígenos de Protozoos/genética , Línea Celular , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Humanos , Hígado/parasitología , Proteínas de la Membrana/genética , Ratones , Ratones SCID , Proteínas Protozoarias/genética , Receptores de Superficie Celular/genética
10.
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
11.
Bioorg Med Chem ; 24(9): 1993-2010, 2016 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-27021426

RESUMEN

The use of arginine isosteres is a known strategy to overcome poor membrane permeability commonly associated with peptides or peptidomimetics that possess this highly polar amino acid. Here, we apply this strategy to peptidomimetics that are potent inhibitors of the malarial protease, plasmepsin V, with the aim of enhancing their activity against Plasmodium parasites, and exploring the structure-activity relationship of the P3 arginine within the S3 pocket of plasmepsin V. Of the arginine isosteres trialled in the P3 position, we discovered that canavanine was the ideal and that this peptidomimetic potently inhibits plasmepsin V, efficiently blocks protein export and inhibits parasite growth. Structure studies of the peptidomimetics bound to plasmepsin V provided insight into the structural basis for the enzyme activity observed in vitro and provides further evidence why plasmepsin V is highly sensitive to substrate modification.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Peptidomiméticos/química , Plasmodium vivax/enzimología , Animales , Espectroscopía de Resonancia Magnética , Espectrometría de Masa por Ionización de Electrospray
12.
Nature ; 463(7281): 627-31, 2010 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-20130643

RESUMEN

Plasmodium falciparum causes the virulent form of malaria and disease manifestations are linked to growth inside infected erythrocytes. To survive and evade host responses the parasite remodels the erythrocyte by exporting several hundred effector proteins beyond the surrounding parasitophorous vacuole membrane. A feature of exported proteins is a pentameric motif (RxLxE/Q/D) that is a substrate for an unknown protease. Here we show that the protein responsible for cleavage of this motif is plasmepsin V (PMV), an aspartic acid protease located in the endoplasmic reticulum. PMV cleavage reveals the export signal (xE/Q/D) at the amino terminus of cargo proteins. Expression of an identical mature protein with xQ at the N terminus generated by signal peptidase was not exported, demonstrating that PMV activity is essential and linked with other key export events. Identification of the protease responsible for export into erythrocytes provides a novel target for therapeutic intervention against this devastating disease.


Asunto(s)
Ácido Aspártico Endopeptidasas/metabolismo , Eritrocitos/metabolismo , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Señales de Clasificación de Proteína , Proteínas Protozoarias/metabolismo , Secuencias de Aminoácidos , Animales , Antimaláricos/farmacología , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/aislamiento & purificación , Retículo Endoplásmico/enzimología , Retículo Endoplásmico/metabolismo , Eritrocitos/citología , Eritrocitos/parasitología , Inhibidores de la Proteasa del VIH/farmacología , Humanos , Malaria Falciparum/metabolismo , Malaria Falciparum/patología , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transporte de Proteínas , Proteínas Protozoarias/química
13.
Mol Cell Proteomics ; 13(5): 1153-64, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24532842

RESUMEN

The routine study of human malaria liver-stage biology in vitro is hampered by low infection efficiency of human hepatocellular carcinoma (HCC) lines (<0.1%), poor understanding of steady-state HCC biology, and lack of appropriate tools for trace sample analysis. HC-04 is the only HCC that supports complete development of human malaria parasites. We hypothesized that HCCs are in various intermediate stages of the epithelial-mesenchymal transition (EMT) and HC-04s retain epithelial characteristics that permit infection. We developed a facile analytical approach to test this hypothesis viz. the HC-04 response to hepatocyte growth factor (HGF). We used online two-dimensional liquid chromatography tandem mass spectrometry (2D-LC-MS/MS) to quantify protein expression profiles in HC-04 pre-/post-HGF treatment and validated these results by RT-qPCR and microscopy. We successfully increased protein identification efficiency over offline-2D methods by 12-fold, using less sample material, allowing robust protein quantification. We observed expected up-regulation and down-regulation of EMT protein markers in response to HGF, but also unexpected cellular responses. We also observed that HC-04 is generally more susceptible to HGF-mediated signaling than what was observed for HepG2, a widely used, but poor malaria liver stage-HCC model. Our analytical approach to understanding the basic biology of HC-04 helps us understand the factors that may influence its utility as a model for malaria liver-stage development. We observed that HC-04 treatment with HGF prior to the addition of Plasmodium falciparum sporozoites did not facilitate cell invasion, which suggests unlinking the effect of HGF on malaria liver stage development from hepatocyte invasion. Finally, our 2D-LC-MS/MS approach and broadly applicable experimental strategy should prove useful in the analysis of various hepatocyte-pathogen interactions, tumor progression, and early disease events.


Asunto(s)
Línea Celular Tumoral/parasitología , Factor de Crecimiento de Hepatocito/metabolismo , Hepatocitos/parasitología , Malaria Falciparum/parasitología , Modelos Biológicos , Plasmodium falciparum/fisiología , Línea Celular Tumoral/citología , Transición Epitelial-Mesenquimal , Perfilación de la Expresión Génica , Células Hep G2 , Humanos , Malaria Falciparum/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Transporte de Proteínas , Proteómica , Proteínas Protozoarias/metabolismo
14.
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
15.
Nature ; 459(7249): 945-9, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19536257

RESUMEN

Several hundred malaria parasite proteins are exported beyond an encasing vacuole and into the cytosol of the host erythrocyte, a process that is central to the virulence and viability of the causative Plasmodium species. The trafficking machinery responsible for this export is unknown. Here we identify in Plasmodium falciparum a translocon of exported proteins (PTEX), which is located in the vacuole membrane. The PTEX complex is ATP-powered, and comprises heat shock protein 101 (HSP101; a ClpA/B-like ATPase from the AAA+ superfamily, of a type commonly associated with protein translocons), a novel protein termed PTEX150 and a known parasite protein, exported protein 2 (EXP2). EXP2 is the potential channel, as it is the membrane-associated component of the core PTEX complex. Two other proteins, a new protein PTEX88 and thioredoxin 2 (TRX2), were also identified as PTEX components. As a common portal for numerous crucial processes, this translocon offers a new avenue for therapeutic intervention.


Asunto(s)
Malaria Falciparum/parasitología , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Animales Modificados Genéticamente , Modelos Biológicos , Unión Proteica , Transporte de Proteínas
16.
J Biol Chem ; 287(45): 38101-9, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22977236

RESUMEN

The mechanism of translocation of RxLR effectors from plant pathogenic oomycetes into the cytoplasm of their host is currently the object of intense research activity and debate. Here, we report the biochemical and thermodynamic characterization of the Phytophthora infestans effector AVR3a in vitro. We show that the amino acids surrounding the RxLR leader mediate homodimerization of the protein. Dimerization was considerably attenuated by a localized mutation within the RxLR motif that was previously described to prevent translocation of the protein into host. Importantly, we confirm that the reported phospholipid-binding properties of AVR3a are mediated by its C-terminal effector domain, not its RxLR leader. However, we show that the observed phospholipid interaction is attributable to a weak association with denatured protein molecules and is therefore most likely physiologically irrelevant.


Asunto(s)
Fosfolípidos/metabolismo , Phytophthora infestans/metabolismo , Multimerización de Proteína , Factores de Virulencia/química , Factores de Virulencia/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Sitios de Unión/genética , Dicroismo Circular , Electroforesis en Gel de Poliacrilamida , Datos de Secuencia Molecular , Mutación , Fosfolípidos/química , Phytophthora infestans/genética , Enfermedades de las Plantas/microbiología , Unión Proteica , Señales de Clasificación de Proteína/genética , Solanum tuberosum/microbiología , Factores de Virulencia/genética
18.
PLoS Pathog ; 6(4): e1000845, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20368977

RESUMEN

Certain environmental microorganisms can cause severe human infections, even in the absence of an obvious requirement for transition through an animal host for replication ("accidental virulence"). To understand this process, we compared eleven isolate genomes of Burkholderia pseudomallei (Bp), a tropical soil microbe and causative agent of the human and animal disease melioidosis. We found evidence for the existence of several new genes in the Bp reference genome, identifying 282 novel genes supported by at least two independent lines of supporting evidence (mRNA transcripts, database homologs, and presence of ribosomal binding sites) and 81 novel genes supported by all three lines. Within the Bp core genome, 211 genes exhibited significant levels of positive selection (4.5%), distributed across many cellular pathways including carbohydrate and secondary metabolism. Functional experiments revealed that certain positively selected genes might enhance mammalian virulence by interacting with host cellular pathways or utilizing host nutrients. Evolutionary modifications improving Bp environmental fitness may thus have indirectly facilitated the ability of Bp to colonize and survive in mammalian hosts. These findings improve our understanding of the pathogenesis of melioidosis, and establish Bp as a model system for studying the genetics of accidental virulence.


Asunto(s)
Evolución Biológica , Burkholderia pseudomallei/genética , Burkholderia pseudomallei/patogenicidad , Genes Bacterianos , Animales , Secuencia de Bases , Femenino , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Genoma Bacteriano , Melioidosis/genética , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Virulencia/genética
19.
Nat Commun ; 13(1): 4400, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906227

RESUMEN

Tryptophan C-mannosylation stabilizes proteins bearing a thrombospondin repeat (TSR) domain in metazoans. Here we show that Plasmodium falciparum expresses a DPY19 tryptophan C-mannosyltransferase in the endoplasmic reticulum and that DPY19-deficiency abolishes C-glycosylation, destabilizes members of the TRAP adhesin family and inhibits transmission to mosquitoes. Imaging P. falciparum gametogenesis in its entirety in four dimensions using lattice light-sheet microscopy reveals defects in ΔDPY19 gametocyte egress and exflagellation. While egress is diminished, ΔDPY19 microgametes still fertilize macrogametes, forming ookinetes, but these are abrogated for mosquito infection. The gametogenesis defects correspond with destabilization of MTRAP, which we show is C-mannosylated in P. falciparum, and the ookinete defect is concordant with defective CTRP secretion on the ΔDPY19 background. Genetic complementation of DPY19 restores ookinete infectivity, sporozoite production and C-mannosylation activity. Therefore, tryptophan C-mannosylation by DPY19 ensures TSR protein quality control at two lifecycle stages for successful transmission of the human malaria parasite.


Asunto(s)
Culicidae , Malaria Falciparum , Animales , Culicidae/metabolismo , Glicosilación , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Trombospondinas/metabolismo , Triptófano/metabolismo
20.
ACS Med Chem Lett ; 13(11): 1745-1754, 2022 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-36385924

RESUMEN

Drug resistance to first-line antimalarials-including artemisinin-is increasing, resulting in a critical need for the discovery of new agents with novel mechanisms of action. In collaboration with the Walter and Eliza Hall Institute and with funding from the Wellcome Trust, a phenotypic screen of Merck's aspartyl protease inhibitor library identified a series of plasmepsin X (PMX) hits that were more potent than chloroquine. Inspired by a PMX homology model, efforts to optimize the potency resulted in the discovery of leads that, in addition to potently inhibiting PMX, also inhibit another essential aspartic protease, plasmepsin IX (PMIX). Further potency and pharmacokinetic profile optimization efforts culminated in the discovery of WM382, a very potent dual PMIX/X inhibitor with robust in vivo efficacy at multiple stages of the malaria parasite life cycle and an excellent resistance profile.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda