Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(1): 105586, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38141766

RESUMEN

About 247 million cases of malaria occurred in 2021 with Plasmodium falciparum accounting for the majority of 619,000 deaths. In the absence of a widely available vaccine, chemotherapy remains crucial to prevent, treat, and contain the disease. The efficacy of several drugs currently used in the clinic is likely to suffer from the emergence of resistant parasites. A global effort to identify lead compounds led to several initiatives such as the Medicine for Malaria Ventures (MMV), a repository of compounds showing promising efficacy in killing the parasite in cell-based assays. Here, we used mass spectrometry coupled with cellular thermal shift assay to identify putative protein targets of MMV000848, a compound with an in vitro EC50 of 0.5 µM against the parasite. Thermal shift assays showed a strong increase of P. falciparum purine nucleoside phosphorylase (PfPNP) melting temperature by up to 15 °C upon incubation with MMV000848. Binding and enzymatic assays returned a KD of 1.52 ± 0.495 µM and an IC50 value of 21.5 ± 2.36 µM. The inhibition is competitive with respect to the substrate, as confirmed by a cocrystal structure of PfPNP bound with MMV000848 at the active site, determined at 1.85 Å resolution. In contrast to transition states inhibitors, MMV000848 specifically inhibits the parasite enzyme but not the human ortholog. An isobologram analysis shows subadditivity with immucillin H and with quinine respectively, suggesting overlapping modes of action between these compounds. These results point to PfPNP as a promising antimalarial target and suggest avenues to improve inhibitor potency.


Asunto(s)
Antimaláricos , Plasmodium falciparum , Purina-Nucleósido Fosforilasa , Antimaláricos/química , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/enzimología , Purina-Nucleósido Fosforilasa/química , Quinina/química , Espectrometría de Masas , Unión Proteica
2.
EMBO Rep ; 24(10): e57090, 2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37592911

RESUMEN

The complex life cycle of the human malaria parasite, Plasmodium falciparum, is driven by specific transcriptional programs, but it is unclear how most genes are activated or silenced at specific times. There is an association between transcription and spatial organization; however, the molecular mechanisms behind genome organization are unclear. While P. falciparum lacks key genome-organizing proteins found in metazoans, it has all core components of the cohesin complex. To investigate the role of cohesin in P. falciparum, we functionally characterize the cohesin subunit Structural Maintenance of Chromosomes protein 3 (SMC3). SMC3 knockdown during early stages of the intraerythrocytic developmental cycle (IDC) upregulates a subset of genes involved in erythrocyte egress and invasion, which are normally expressed at later stages. ChIP-seq analyses reveal that during the IDC, SMC3 enrichment at the promoter regions of these genes inversely correlates with gene expression and chromatin accessibility. These data suggest that SMC3 binding contributes to the repression of specific genes until their appropriate time of expression, revealing a new mode of stage-specific gene repression in P. falciparum.

3.
Cell Microbiol ; 23(2): e13277, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33040440

RESUMEN

About half the world's population is at risk of malaria, with Plasmodium falciparum malaria being responsible for the most malaria related deaths globally. Antimalarial drugs such as chloroquine and artemisinin are directed towards the proliferating intra-erythrocytic stages of the parasite, which is responsible for all the clinical symptoms of the disease. These antimalarial drugs have been reported to function via multiple pathways, one of which induces DNA damage via the generation of free radicals and reactive oxygen species. An urgent need to understand the mechanistic details of drug response and resistance is highlighted by the decreasing clinical efficacy of the front line drug, Artemisinin. The replication factor C subunit 1 is an important component of the DNA replication machinery and DNA damage response mechanism. Here we show the translocation of PfRFC1 from an intranuclear localisation to the nuclear periphery, indicating an orchestrated progression of distinct patterns of replication in the developing parasites. PfRFC1 responds to genotoxic stress via elevated protein levels in soluble and chromatin bound fractions. Reduction of PfRFC1 protein levels upon treatment with antimalarials suggests an interplay of replication, apoptosis and DNA repair pathways leading to cell death. Additionally, mislocalisation of the endogenously tagged protein confirmed its essential role in parasites' replication and DNA repair. This study provides key insights into DNA replication, DNA damage response and cell death in P. falciparum.


Asunto(s)
Antimaláricos/farmacología , Daño del ADN , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/fisiología , Proteína de Replicación C/fisiología , Artesunato/farmacología , Muerte Celular , Cloroquina/farmacología , Reparación del ADN , Replicación del ADN , ADN Protozoario , Eritrocitos/parasitología , Regulación de la Expresión Génica , Interacciones Huésped-Parásitos , Humanos , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Proteínas Protozoarias/fisiología , Especies Reactivas de Oxígeno/metabolismo
4.
Nucleic Acids Res ; 48(1): 184-199, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31777939

RESUMEN

DNA cytosine modifications are key epigenetic regulators of cellular processes in mammalian cells, with their misregulation leading to varied disease states. In the human malaria parasite Plasmodium falciparum, a unicellular eukaryotic pathogen, little is known about the predominant cytosine modifications, cytosine methylation (5mC) and hydroxymethylation (5hmC). Here, we report the first identification of a hydroxymethylcytosine-like (5hmC-like) modification in P. falciparum asexual blood stages using a suite of biochemical methods. In contrast to mammalian cells, we report 5hmC-like levels in the P. falciparum genome of 0.2-0.4%, which are significantly higher than the methylated cytosine (mC) levels of 0.01-0.05%. Immunoprecipitation of hydroxymethylated DNA followed by next generation sequencing (hmeDIP-seq) revealed that 5hmC-like modifications are enriched in gene bodies with minimal dynamic changes during asexual development. Moreover, levels of the 5hmC-like base in gene bodies positively correlated to transcript levels, with more than 2000 genes stably marked with this modification throughout asexual development. Our work highlights the existence of a new predominant cytosine DNA modification pathway in P. falciparum and opens up exciting avenues for gene regulation research and the development of antimalarials.


Asunto(s)
5-Metilcitosina/análogos & derivados , ADN Protozoario/genética , Epigénesis Genética , Genoma de Protozoos , Plasmodium falciparum/genética , ARN Mensajero/genética , 5-Metilcitosina/metabolismo , Citosina/metabolismo , Metilación de ADN , ADN Protozoario/metabolismo , Eritrocitos/parasitología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hidroxilación , Plasmodium falciparum/metabolismo , ARN Mensajero/metabolismo
5.
Mol Syst Biol ; 16(8): e9569, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32816370

RESUMEN

Mutually exclusive expression of the var multigene family is key to immune evasion and pathogenesis in Plasmodium falciparum, but few factors have been shown to play a direct role. We adapted a CRISPR-based proteomics approach to identify novel factors associated with var genes in their natural chromatin context. Catalytically inactive Cas9 ("dCas9") was targeted to var gene regulatory elements, immunoprecipitated, and analyzed with mass spectrometry. Known and novel factors were enriched including structural proteins, DNA helicases, and chromatin remodelers. Functional characterization of PfISWI, an evolutionarily divergent putative chromatin remodeler enriched at the var gene promoter, revealed a role in transcriptional activation. Proteomics of PfISWI identified several proteins enriched at the var gene promoter such as acetyl-CoA synthetase, a putative MORC protein, and an ApiAP2 transcription factor. These findings validate the CRISPR/dCas9 proteomics method and define a new var gene-associated chromatin complex. This study establishes a tool for targeted chromatin purification of unaltered genomic loci and identifies novel chromatin-associated factors potentially involved in transcriptional control and/or chromatin organization of virulence genes in the human malaria parasite.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Plasmodium falciparum/patogenicidad , Proteómica/métodos , Factores de Transcripción/metabolismo , Factores de Virulencia/genética , Animales , Antígenos de Protozoos/genética , Antígenos de Protozoos/metabolismo , Sistemas CRISPR-Cas , Secuenciación de Inmunoprecipitación de Cromatina , Humanos , Intrones , Espectrometría de Masas , Plasmodium falciparum/genética , Plasmodium falciparum/inmunología , Regiones Promotoras Genéticas , Mapas de Interacción de Proteínas , Factores de Virulencia/metabolismo
6.
Cell Microbiol ; 22(9): e13232, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32452132

RESUMEN

Plasmodium falciparum responsible for the most virulent form of malaria invades human erythrocytes through multiple ligand-receptor interactions. The P. falciparum reticulocyte binding protein homologues (PfRHs) are expressed at the apical end of merozoites and form interactions with distinct erythrocyte surface receptors that are important for invasion. Here using a range of monoclonal antibodies (mAbs) against different regions of PfRH1 we have investigated the role of PfRH processing during merozoite invasion. We show that PfRH1 gets differentially processed during merozoite maturation and invasion and provide evidence that the different PfRH1 processing products have distinct functions during invasion. Using in-situ Proximity Ligation and FRET assays that allow probing of interactions at the nanometre level we show that a subset of PfRH1 products form close association with micronemal proteins Apical Membrane Antigen 1 (AMA1) in the moving junction suggesting a critical role in facilitating junction formation and active invasion. Our data provides evidence that time dependent processing of PfRH proteins is a mechanism by which the parasite is able to regulate distinct functional activities of these large processes. The identification of a specific close association with AMA1 in the junction now may also provide new avenues to target these interactions to prevent merozoite invasion.


Asunto(s)
Antígenos de Protozoos/metabolismo , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Proteínas de la Membrana/metabolismo , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Reticulocitos/metabolismo , Uniones Estrechas/metabolismo , Anticuerpos Monoclonales , Antígenos de Protozoos/genética , Eritrocitos/metabolismo , Proteínas de la Membrana/genética , Merozoítos/metabolismo , Plasmodium falciparum/química , Proteínas Protozoarias/genética , Uniones Estrechas/parasitología
7.
Mol Cell Proteomics ; 18(5): 837-853, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30718293

RESUMEN

Efforts to develop vaccines against malaria represent a major research target. The observations that 1) sterile protection can be obtained when the host is exposed to live parasites and 2) the immunity against blood stage parasite is principally mediated by protective antibodies suggest that a protective vaccine is feasible. However, only a small number of proteins have been investigated so far and most of the Plasmodium proteome has yet to be explored. To date, only few immunodominant antigens have emerged for testing in clinical trials but no formulation has led to substantial protection in humans. The nature of parasite molecules associated with protection remains elusive. Here, immunomic screening of mice immune sera with different protection efficiencies against the whole parasite proteome allowed us to identify a large repertoire of antigens validated by screening a library expressing antigens. The calculation of weighted scores reflecting the likelihood of protection of each antigen using five predictive criteria derived from immunomic and proteomic data sets, highlighted a priority list of protective antigens. Altogether, the approach sheds light on conserved antigens across Plasmodium that are amenable to targeting by the host immune system upon merozoite invasion and blood stage development. Most of these antigens have preliminary protection data but have not been widely considered as candidate for vaccine trials, opening new perspectives that overcome the limited choice of immunodominant, poorly protective vaccines currently being the focus of malaria vaccine researches.


Asunto(s)
Antígenos de Protozoos/inmunología , Malaria/inmunología , Malaria/prevención & control , Animales , Antígenos de Protozoos/química , Células CHO , Cricetinae , Cricetulus , Membrana Eritrocítica/metabolismo , Sueros Inmunes , Malaria/sangre , Merozoítos/crecimiento & desarrollo , Merozoítos/inmunología , Ratones Endogámicos BALB C , Parásitos/crecimiento & desarrollo , Plasmodium/crecimiento & desarrollo , Plasmodium/inmunología , Desnaturalización Proteica , Dominios Proteicos , Proteómica , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Reproducibilidad de los Resultados
8.
Artículo en Inglés | MEDLINE | ID: mdl-32071059

RESUMEN

We report a systematic, cellular phenotype-based antimalarial screening of the Medicines for Malaria Venture Pathogen Box collection, which facilitated the identification of specific blockers of late-stage intraerythrocytic development of Plasmodium falciparum First, from standard growth inhibition assays, we identified 173 molecules with antimalarial activity (50% effective concentration [EC50] ≤ 10 µM), which included 62 additional molecules over previously known antimalarial candidates from the Pathogen Box. We identified 90 molecules with EC50 of ≤1 µM, which had significant effect on the ring-trophozoite transition, while 9 molecules inhibited the trophozoite-schizont transition and 21 molecules inhibited the schizont-ring transition (with ≥50% parasites failing to proceed to the next stage) at 1 µM. We therefore rescreened all 173 molecules and validated hits in microscopy to prioritize 12 hits as selective blockers of the schizont-ring transition. Seven of these molecules inhibited the calcium ionophore-induced egress of Toxoplasma gondii, a related apicomplexan parasite, suggesting that the inhibitors may be acting via a conserved mechanism which could be further exploited for target identification studies. We demonstrate that two molecules, MMV020670 and MMV026356, identified as schizont inhibitors in our screens, induce the fragmentation of DNA in merozoites, thereby impairing their ability to egress and invade. Further mechanistic studies would facilitate the therapeutic exploitation of these molecules as broadly active inhibitors targeting late-stage development and egress of apicomplexan parasites relevant to human health.


Asunto(s)
Antimaláricos/farmacología , Evaluación Preclínica de Medicamentos/métodos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Fragmentación del ADN/efectos de los fármacos , Humanos , Merozoítos/efectos de los fármacos , Pruebas de Sensibilidad Parasitaria , Esquizontes/efectos de los fármacos , Trofozoítos/efectos de los fármacos
9.
PLoS Pathog ; 14(10): e1007298, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30286211

RESUMEN

Natural killer (NK) cells provide the first line of defense against malaria parasite infection. However, the molecular mechanisms through which NK cells are activated by parasites are largely unknown, so is the molecular basis underlying the variation in NK cell responses to malaria infection in the human population. Here, we compared transcriptional profiles of responding and non-responding NK cells following exposure to Plasmodium-infected red blood cells (iRBCs) and identified MDA5, a RIG-I-like receptor involved in sensing cytosolic RNAs, to be differentially expressed. Knockout of MDA5 in responding human NK cells by CRISPR/cas9 abolished NK cell activation, IFN-γ secretion, lysis of iRBCs. Similarly, inhibition of TBK1/IKKε, an effector molecule downstream of MDA5, also inhibited activation of responding NK cells. Conversely, activation of MDA5 by liposome-packaged poly I:C restored non-responding NK cells to lyse iRBCs. We further show that microvesicles containing large parasite RNAs from iRBCs activated NK cells by fusing with NK cells. These findings suggest that NK cells are activated through the MDA5 pathway by parasite RNAs that are delivered to the cytoplasm of NK cells by microvesicles from iRBCs. The difference in MDA5 expression between responding and non-responding NK cells following exposure to iRBCs likely contributes to the variation in NK cell responses to malaria infection in the human population.


Asunto(s)
Micropartículas Derivadas de Células/inmunología , Eritrocitos/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Células Asesinas Naturales/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Sistemas CRISPR-Cas , Células Cultivadas , Citoplasma/metabolismo , Eritrocitos/metabolismo , Eritrocitos/parasitología , Humanos , Helicasa Inducida por Interferón IFIH1/antagonistas & inhibidores , Helicasa Inducida por Interferón IFIH1/genética , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/parasitología , Activación de Linfocitos , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Plasmodium falciparum/aislamiento & purificación
10.
Mol Syst Biol ; 14(10): e8009, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30287681

RESUMEN

Among components of the translational machinery, ribonucleoside modifications on tRNAs are emerging as critical regulators of cell physiology and stress response. Here, we demonstrate highly coordinated behavior of the repertoire of tRNA modifications of Plasmodium falciparum throughout the intra-erythrocytic developmental cycle (IDC). We observed both a synchronized increase in 22 of 28 modifications from ring to trophozoite stage, consistent with tRNA maturation during translational up-regulation, and asynchronous changes in six modifications. Quantitative analysis of ~2,100 proteins across the IDC revealed that up- and down-regulated proteins in late but not early stages have a marked codon bias that directly correlates with parallel changes in tRNA modifications and enhanced translational efficiency. We thus propose a model in which tRNA modifications modulate the abundance of stage-specific proteins by enhancing translation efficiency of codon-biased transcripts for critical genes. These findings reveal novel epitranscriptomic and translational control mechanisms in the development and pathogenesis of Plasmodium parasites.


Asunto(s)
Plasmodium falciparum/fisiología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , ARN de Transferencia/metabolismo , Codón , Epigénesis Genética , Eritrocitos , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Plasmodium falciparum/genética , Biosíntesis de Proteínas , Procesamiento Proteico-Postraduccional , Proteómica/métodos
11.
Cell Microbiol ; 19(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28030753

RESUMEN

The extensive modification of Plasmodium falciparum-infected erythrocytes by variant surface antigens plays a major role in immune evasion and malaria-induced pathology. Here, using high-resolution microscopy, we visualize the spatio-temporal expression dynamics of STEVOR, an important variant surface antigens family, in a stage-dependent manner. We demonstrate that it is exported to the cell surface where protein molecules cluster and preferentially localize in proximity to knobs. Quantitative evidence from our force measurements and microfluidic assays reveal that STEVOR can effectively mediate the formation of stable, robust rosettes under static and physiologically relevant flow conditions. Our results extend previously published studies in P. falciparum and emphasize the role of STEVOR in rosetting, an important contributor to disease pathology.


Asunto(s)
Antígenos de Protozoos/genética , Antígenos de Superficie/genética , Adhesión Celular/genética , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/genética , Antígenos de Protozoos/biosíntesis , Antígenos de Superficie/biosíntesis , Adhesión Celular/fisiología , Línea Celular , Eritrocitos/parasitología , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/biosíntesis , Formación de Roseta
12.
Cell Microbiol ; 19(9)2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28409866

RESUMEN

The successful invasion of Plasmodium is an essential step in their life cycle. The parasite reticulocyte-binding protein homologues (RHs) and erythrocyte-binding like proteins are two families involved in the invasion leading to merozoite-red blood cell (RBC) junction formation. Ca2+ signaling has been shown to play a critical role in the invasion. RHs have been linked to Ca2+ signaling, which triggers the erythrocyte-binding like proteins release ahead of junction formation, consistent with RHs performing an initial sensing function in identifying suitable RBCs. RH5, the only essential RHs, is a highly promising vaccine candidate. RH5-basigin interaction is essential for merozoite invasion and also important in determining host tropism. Here, we show that RH5 has a distinct function from the other RHs. We show that RH5-Basigin interaction on its own triggers a Ca2+ signal in the RBC resulting in changes in RBC cytoskeletal proteins phosphorylation and overall alterations in RBC cytoskeleton architecture. Antibodies targeting RH5 that block the signal prevent invasion before junction formation consistent with the Ca2+ signal in the RBC leading to rearrangement of the cytoskeleton required for invasion. This work provides the first time a functional context for the essential role of RH5 and will now open up new avenues to target merozoite invasion.


Asunto(s)
Basigina/metabolismo , Señalización del Calcio/fisiología , Proteínas Portadoras/metabolismo , Eritrocitos/fisiología , Merozoítos/patogenicidad , Plasmodium falciparum/patogenicidad , Anticuerpos Monoclonales/inmunología , Antígenos de Protozoos/biosíntesis , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/inmunología , Línea Celular , Citoesqueleto/parasitología , Citoesqueleto/patología , Eritrocitos/parasitología , Interacciones Huésped-Parásitos/fisiología , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/biosíntesis
13.
Mol Microbiol ; 102(3): 386-404, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27438226

RESUMEN

Erythrocyte invasion by merozoite is a multistep process involving multiple ligand-receptor interactions. The Plasmodium falciparum reticulocyte binding protein homologues (PfRHs) consists of five functional members. The differential expression of PfRHs has been linked to the utilization of different invasion pathways by the merozoites as well as a mechanism of immune evasion. PfRHs are expressed at the apical end of merozoite and form interactions with distinct red blood cell (RBC) surface receptors that are important for successful invasion. Here we show that PfRH2b undergoes processing before and during merozoite invasion. The different processed fragments bind to chymotrypsin sensitive RBC surface receptors. We also show that PfRH2b follows the merozoite tight junction during invasion. Monoclonal antibodies (mAbs) inhibit merozoites invasion by blocking tight junction formation. mAbs binding to PfRH2b block merozoites intracellular Ca2+ signal necessary for EBA175 surface expression. The data suggests that a conserved function of PfRHs, where their interaction with RBC surface receptors facilitated recruitment of EBA175 and other tight junction proteins necessary for merozoite invasion by modulating merozoite intracellular Ca2+ signals.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Eritrocitos/parasitología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Quimotripsina/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Interacciones Huésped-Parásitos , Humanos , Merozoítos/efectos de los fármacos , Merozoítos/metabolismo , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/metabolismo , Receptores de Superficie Celular/metabolismo
14.
Cell Microbiol ; 18(12): 1739-1750, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27130708

RESUMEN

The development of an effective malaria vaccine has remained elusive even until today. This is because of our incomplete understanding of the immune mechanisms that confer and/or correlate with protection. Human volunteers have been protected experimentally from a subsequent challenge by immunization with Plasmodium falciparum sporozoites under drug cover. Here, we demonstrate that sera from the protected individuals contain neutralizing antibodies against the pre-erythrocytic stage. To identify the antigen(s) recognized by these antibodies, a newly developed library of P. falciparum antigens was screened with the neutralizing sera. Antibodies from protected individuals recognized a broad antigenic repertoire of which three antigens, PfMAEBL, PfTRAP and PfSEA1 were recognized by most protected individuals. As a proof of principle, we demonstrated that anti-PfMAEBL antibodies block liver stage development in human hepatocytes. Thus, these antigens identified are promising targets for vaccine development against malaria.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Antígenos de Protozoos/inmunología , Inmunidad Humoral , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología , Receptores de Superficie Celular/inmunología , Animales , Anticuerpos Neutralizantes/biosíntesis , Antígenos de Protozoos/genética , Antimaláricos/uso terapéutico , Cloroquina/uso terapéutico , Reacciones Cruzadas , Expresión Génica , Hepatocitos/efectos de los fármacos , Hepatocitos/inmunología , Hepatocitos/parasitología , Humanos , Sueros Inmunes/química , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Biblioteca de Péptidos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Receptores de Superficie Celular/genética , Esporozoítos/inmunología , Vacunación
15.
Proc Natl Acad Sci U S A ; 111(4): 1479-84, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24474774

RESUMEN

Immunodeficient mouse-human chimeras provide a powerful approach to study host-specific pathogens, such as Plasmodium falciparum that causes human malaria. Supplementation of immunodeficient mice with human RBCs supports infection by human Plasmodium parasites, but these mice lack the human immune system. By combining human RBC supplementation and humanized mice that are optimized for human immune cell reconstitution, we have developed RBC-supplemented, immune cell-optimized humanized (RICH) mice that support multiple cycles of P. falciparum infection. Depletion of human natural killer (NK) cells, but not macrophages, in RICH mice results in a significant increase in parasitemia. Further studies in vitro show that NK cells preferentially interact with infected RBCs (iRBCs), resulting in the activation of NK cells and the elimination of iRBCs in a contact-dependent manner. We show that the adhesion molecule lymphocyte-associated antigen 1 is required for NK cell interaction with and elimination of iRBCs. Development of RICH mice and validation of P. falciparum infection should facilitate the dissection of human immune responses to malaria parasite infection and the evaluation of therapeutics and vaccines.


Asunto(s)
Eritrocitos/parasitología , Células Asesinas Naturales/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Animales , Adhesión Celular , Humanos , Malaria Falciparum/sangre , Ratones , Parasitemia/inmunología
16.
Blood ; 123(18): e100-9, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24652986

RESUMEN

Rosetting phenomenon has been linked to malaria pathogenesis. Although rosetting occurs in all causes of human malaria, most data on this subject has been derived from Plasmodium falciparum. Here, we investigate the function and factors affecting rosette formation in Plasmodium vivax. To achieve this, we used a range of novel ex vivo protocols to study fresh and cryopreserved P vivax (n = 135) and P falciparum (n = 77) isolates from Thailand. Rosetting is more common in vivax than falciparum malaria, both in terms of incidence in patient samples and percentage of infected erythrocytes forming rosettes. Rosetting to P vivax asexual and sexual stages was evident 20 hours postreticulocyte invasion, reaching a plateau after 30 hours. Host ABO blood group, reticulocyte count, and parasitemia were not correlated with P vivax rosetting. Importantly, mature erythrocytes (normocytes), rather than reticulocytes, preferentially form rosetting complexes, indicating that this process is unlikely to directly facilitate merozoite invasion. Although antibodies against host erythrocyte receptors CD235a and CD35 had no effect, Ag-binding fragment against the BRIC 4 region of CD236R significantly inhibited rosette formation. Rosetting assays using CD236R knockdown normocytes derived from hematopoietic stem cells further supports the role of glycophorin C as a receptor in P vivax rosette formation.


Asunto(s)
Eritrocitos/metabolismo , Eritrocitos/parasitología , Glicoforinas/metabolismo , Malaria Vivax/metabolismo , Plasmodium vivax/inmunología , Formación de Roseta/métodos , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Criopreservación/métodos , Eritrocitos/patología , Técnicas de Silenciamiento del Gen , Glicoforinas/genética , Glicoforinas/inmunología , Humanos , Malaria Vivax/diagnóstico , Malaria Vivax/parasitología , Plasmodium vivax/aislamiento & purificación , Receptores de Complemento 3b/antagonistas & inhibidores , Flujo de Trabajo
17.
Mol Microbiol ; 91(5): 918-34, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24372851

RESUMEN

Drug resistance in Plasmodium falciparum remains a challenge for the malaria eradication programmes around the world. With the emergence of artemisinin resistance, the efficacy of the partner drugs in the artemisinin combination therapies (ACT) that include quinoline-based drugs is becoming critical. So far only few resistance markers have been identified from which only two transmembrane transporters namely PfMDR1 (an ATP-binding cassette transporter) and PfCRT (a drug-metabolite transporter) have been experimentally verified. Another P. falciparum transporter, the ATP-binding cassette containing multidrug resistance-associated protein (PfMRP2) represents an additional possible factor of drug resistance in P. falciparum. In this study, we identified a parasite clone that is derived from the 3D7 P. falciparum strain and shows increased resistance to chloroquine, mefloquine and quinine through the trophozoite and schizont stages. We demonstrate that the resistance phenotype is caused by a 4.1 kb deletion in the 5' upstream region of the pfmrp2 gene that leads to an alteration in the pfmrp2 transcription and thus increased level of PfMRP2 protein. These results also suggest the importance of putative promoter elements in regulation of gene expression during the P. falciparum intra-erythrocytic developmental cycle and the potential of genetic polymorphisms within these regions to underlie drug resistance.


Asunto(s)
Resistencia a Medicamentos/genética , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Polimorfismo Genético , Regiones Promotoras Genéticas/genética , Proteínas Protozoarias/genética , Quinolinas/farmacología , Antimaláricos/farmacología , Emparejamiento Base/genética , Secuencia de Bases , Células Clonales , Resistencia a Medicamentos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genoma de Protozoos/genética , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Transporte de Proteínas/efectos de los fármacos , Proteínas Protozoarias/metabolismo , Análisis de Secuencia de ADN , Eliminación de Secuencia/genética , Transcripción Genética/efectos de los fármacos , Transcriptoma/genética
18.
Genome Res ; 22(5): 925-38, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22415456

RESUMEN

Malaria genetic variation has been extensively characterized, but the level of epigenetic plasticity remains largely unexplored. Here we provide a comprehensive characterization of transcriptional variation in the most lethal malaria parasite, Plasmodium falciparum, based on highly accurate transcriptional analysis of isogenic parasite lines grown under homogeneous conditions. This analysis revealed extensive transcriptional heterogeneity within genetically homogeneous clonal parasite populations. We show that clonally variant expression controlled at the epigenetic level is an intrinsic property of specific genes and gene families, the majority of which participate in host-parasite interactions. Intrinsic transcriptional variability is not restricted to genes involved in immune evasion, but also affects genes linked to lipid metabolism, protein folding, erythrocyte remodeling, or transcriptional regulation, among others, indicating that epigenetic variation results in both antigenic and functional variation. We observed a general association between heterochromatin marks and clonally variant expression, extending previous observations for specific genes to essentially all variantly expressed gene families. These results suggest that phenotypic variation of functionally unrelated P. falciparum gene families is mediated by a common mechanism based on reversible formation of H3K9me3-based heterochromatin. In changing environments, diversity confers fitness to a population. Our results support the idea that P. falciparum uses a bet-hedging strategy, as an alternative to directed transcriptional responses, to adapt to common fluctuations in its environment. Consistent with this idea, we found that transcriptionally different isogenic parasite lines markedly differed in their survival to heat-shock mimicking febrile episodes and adapted to periodic heat-shock with a pattern consistent with natural selection of pre-existing parasites.


Asunto(s)
Epigénesis Genética , Genes Protozoarios , Plasmodium falciparum/genética , Transcriptoma , Adaptación Fisiológica/genética , Técnicas de Cultivo , Perfilación de la Expresión Génica , Respuesta al Choque Térmico , Heterocromatina/metabolismo , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/fisiología , Transcripción Genética , Trofozoítos/crecimiento & desarrollo , Trofozoítos/metabolismo , Trofozoítos/fisiología
19.
Cell Microbiol ; 16(5): 673-86, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24636637

RESUMEN

Development of the erythrocytic malaria parasite requires targeting of parasite proteins into multiple compartments located within and beyond the parasite confine. Beyond the PEXEL/VTS pathway and its characterized players, increasing amount of evidence has highlighted the existence of proteins exported using alternative export-signal(s)/pathway(s); hence, the exportomes currently predicted are incomplete. The nature of these exported proteins which could have a prominent role in most of the Plasmodium species remains elusive. Using P. yoelii variant proteins, we identified a signal associated to lipophilic region that mediates export of P. yoelii proteins. This non-PEXEL signal termed PLASMED is defined by semi-conserved residues and possibly a secondary structure. In vivo characterization of exported-proteins indicated that PLASMED is a bona fide export-signal that allowed us to identify an unseen P. yoelii exportome. The repertoire of the newly predicted exported proteins opens up perspectives for unravelling the remodelling of the host-cell by the parasite, against which new therapies could be elaborated.


Asunto(s)
Plasmodium yoelii/genética , Plasmodium yoelii/metabolismo , Señales de Clasificación de Proteína , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Conformación Proteica , Transporte de Proteínas , Proteínas Protozoarias/química , Proteínas Protozoarias/genética
20.
Malar J ; 14: 110, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25880967

RESUMEN

BACKGROUND: Plasmodium knowlesi is one of five Plasmodium species known to cause malaria in humans and can result in severe illness and death. While a zoonosis in humans, this simian malaria parasite species infects macaque monkeys and serves as an experimental model for in vivo, ex vivo and in vitro studies. It has underpinned malaria discoveries relating to host-pathogen interactions, the immune response and immune evasion strategies. This study investigated differences in P. knowlesi gene expression in samples from ex vivo and in vitro cultures. METHODS: Gene expression profiles were generated using microarrays to compare the stage-specific transcripts detected for a clone of P. knowlesi propagated in the blood of a rhesus macaque host and then grown in an ex-vivo culture, and the same clone adapted to long-term in vitro culture. Parasite samples covering one blood-stage cycle were analysed at four-hour intervals. cDNA was generated and hybridized to an oligoarray representing the P. knowlesi genome. Two replicate experiments were developed from in vitro cultures. Expression values were filtered, normalized, and analysed using R and Perl language and applied to a sine wave model to determine changes in equilibrium and amplitude. Differentially expressed genes from ex vivo and in vitro time points were detected using limma R/Bioconductor and gene set enrichment analysis (GSEA). RESULTS: Major differences were noted between the ex vivo and in vitro time courses in overall gene expression and the length of the cycle (25.5 hours ex vivo; 33.5 hours in vitro). GSEA of genes up-regulated ex vivo showed an enrichment of various genes including SICAvar, ribosomal- associated and histone acetylation pathway genes. In contrast, certain genes involved in metabolism and cell growth, such as porphobilinogen deaminase and tyrosine phosphatase, and one SICAvar gene, were significantly up-regulated in vitro. CONCLUSIONS: This study demonstrates how gene expression in P. knowlesi blood-stage parasites can differ dramatically depending on whether the parasites are grown in vivo, with only one cycle of development ex vivo, or as an adapted isolate in long-term in vitro culture. These data bring emphasis to the importance of studying the parasite, its biology and disease manifestations in the context of the host.


Asunto(s)
Eritrocitos/parasitología , Interacciones Huésped-Parásitos/genética , Plasmodium knowlesi/genética , Plasmodium knowlesi/patogenicidad , Proteínas Protozoarias , Animales , ADN Protozoario/genética , ADN Protozoario/metabolismo , Expresión Génica/genética , Perfilación de la Expresión Génica , Macaca mulatta , Análisis de Secuencia por Matrices de Oligonucleótidos , Plasmodium knowlesi/metabolismo , Proteínas Protozoarias/análisis , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA