Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
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.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
Infect Immun ; 82(6): 2532-41, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24686065

RESUMEN

Recent experimental and clinical studies suggest a crucial role of mechanical splenic filtration in the host's defense against malaria parasites. Subtle changes in red blood cell (RBC) deformability, caused by infection or drug treatment, could influence the pathophysiological outcome. However, in vitro deformability measurements have not been directly linked in vivo with the splenic clearance of RBCs. In this study, mice infected with malaria-inducing Plasmodium yoelii revealed that chloroquine treatment could lead to significant alterations to RBC deformability and increase clearance of both infected and uninfected RBCs in vivo. These results have clear implications for the mechanism of human malarial anemia, a severe pathological condition affecting malaria patients.


Asunto(s)
Deformación Eritrocítica/fisiología , Malaria/fisiopatología , Malaria/parasitología , Plasmodium yoelii , Bazo/fisiopatología , Anemia , Animales , Antimaláricos/farmacología , Cloroquina/farmacología , Modelos Animales de Enfermedad , Femenino , Hemoglobinas/análisis , Masculino , Ratones , Ratones Endogámicos BALB C , Microcirculación/efectos de los fármacos , Bazo/efectos de los fármacos
16.
Blood ; 119(24): e172-80, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22517905

RESUMEN

Achievement of malaria elimination requires development of novel strategies interfering with parasite transmission, including targeting the parasite sexual stages (gametocytes). The formation of Plasmodium falciparum gametocytes in the human host takes several days during which immature gametocyte-infected erythrocytes (GIEs) sequester in host tissues. Only mature stage GIEs circulate in the peripheral blood, available to uptake by the Anopheles vector. Mechanisms underlying GIE sequestration and release in circulation are virtually unknown. We show here that mature GIEs are more deformable than immature stages using ektacytometry and microsphiltration methods, and that a switch in cellular deformability in the transition from immature to mature gametocytes is accompanied by the deassociation of parasite-derived STEVOR proteins from the infected erythrocyte membrane. We hypothesize that mechanical retention contributes to sequestration of immature GIEs and that regained deformability of mature gametocytes is associated with their release in the bloodstream and ability to circulate. These processes are proposed to play a key role in P falciparum gametocyte development in the host and to represent novel and unconventional targets for interfering with parasite transmission.


Asunto(s)
Deformación Eritrocítica/fisiología , Eritrocitos/parasitología , Estadios del Ciclo de Vida , Malaria Falciparum/sangre , Malaria Falciparum/transmisión , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/fisiología , Adulto , Animales , Antígenos de Protozoos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/ultraestructura , Transporte de Proteínas
17.
Cell Microbiol ; 15(1): 35-44, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23046317

RESUMEN

Malaria remains a serious public health problem with significant morbidity and mortality accounting for nearly 20% of all childhood deaths in Africa. The cyclical invasion, cytoadherence and destruction of the host's erythrocyte by the parasite are responsible for the observed disease pathology. The invasive form of the parasite, the merozoite, uses a complex set of interactions between parasite ligands and erythrocyte receptors that leads to the formation of a tight junction and ultimately successful erythrocyte invasion. Understanding the molecular mechanism underlying host cell recognition and invasion is crucial for the development of a targeted intervention strategy. Two parasite protein families termed reticulocyte-binding-like protein homologues (RBL) and the erythrocyte-binding-like (EBL) protein family are conserved in all Plasmodium species and have been shown to play an important role in host cell recognition and invasion. Over the last few years significant new insights have been gained in understanding the function of the RBL family and this review attempts to provide an update with a specific focus on the role of RBL in signal transduction pathways during invasion.


Asunto(s)
Endocitosis , Eritrocitos/parasitología , Plasmodium/patogenicidad , Proteínas Protozoarias/metabolismo , Factores de Virulencia/metabolismo , Modelos Biológicos , Transducción de Señal
18.
Nat Microbiol ; 9(6): 1483-1498, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38632343

RESUMEN

Plasmodium falciparum artemisinin (ART) resistance is driven by mutations in kelch-like protein 13 (PfK13). Quiescence, a key aspect of resistance, may also be regulated by a yet unidentified epigenetic pathway. Transfer RNA modification reprogramming and codon bias translation is a conserved epitranscriptomic translational control mechanism that allows cells to rapidly respond to stress. We report a role for this mechanism in ART-resistant parasites by combining tRNA modification, proteomic and codon usage analyses in ring-stage ART-sensitive and ART-resistant parasites in response to drug. Post-drug, ART-resistant parasites differentially hypomodify mcm5s2U on tRNA and possess a subset of proteins, including PfK13, that are regulated by Lys codon-biased translation. Conditional knockdown of the terminal s2U thiouridylase, PfMnmA, in an ART-sensitive parasite background led to increased ART survival, suggesting that hypomodification can alter the parasite ART response. This study describes an epitranscriptomic pathway via tRNA s2U reprogramming that ART-resistant parasites may employ to survive ART-induced stress.


Asunto(s)
Antimaláricos , Artemisininas , Resistencia a Medicamentos , Plasmodium falciparum , Proteínas Protozoarias , ARN de Transferencia , Plasmodium falciparum/genética , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Resistencia a Medicamentos/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Artemisininas/farmacología , Antimaláricos/farmacología , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Humanos , Malaria Falciparum/parasitología , Proteómica , Codón/genética
19.
Mol Microbiol ; 84(2): 243-59, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22435676

RESUMEN

The Plasmodium falciparum genome is equipped with several subtelomeric gene families that are implicated in parasite virulence and immune evasion. Members of these families are uniformly positioned within heterochromatic domains and are thus subject to variegated expression. The best-studied example is that of the var family encoding the major parasite virulence factor P. falciparum erythrocyte membrane protein 1 (PfEMP1). PfEMP1 undergoes antigenic variation through switches in mutually exclusive var gene transcription. var promoters function as crucial regulatory elements in the underlying epigenetic control strategy. Here, we analysed promoters of upsA, upsB and upsC var, rifA1-type rif, stevor, phist and pfmc-2tm genes and investigated their role in endogenous gene transcription by comparative genome-wide expression profiling of transgenic parasite lines. We find that the three major var promoter types are functionally equal and play an essential role in singular gene choice. Unlike var promoters, promoters of non-var families are not silenced by default, and transcription of non-var families is not subject to the same mode of mutually exclusive transcription as has been observed for var genes. Our findings identified a differential logic in the regulation of var and other subtelomeric virulence gene families, which will have important implications for our understanding and future analyses of phenotypic variation in malaria parasites.


Asunto(s)
Regulación de la Expresión Génica , Genes Protozoarios , Plasmodium falciparum/genética , Plasmodium falciparum/patogenicidad , Factores de Virulencia/biosíntesis , Factores de Virulencia/genética , Perfilación de la Expresión Génica , Regiones Promotoras Genéticas , Transcripción Genética
20.
Cell Rep ; 42(11): 113419, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37952150

RESUMEN

Plasmodium parasites contribute to one of the highest global infectious disease burdens. To achieve this success, the parasite has evolved a range of specialized subcellular compartments to extensively remodel the host cell for its survival. The information to fully understand these compartments is likely hidden in the so far poorly characterized Plasmodium species spatial proteome. To address this question, we determined the steady-state subcellular location of more than 12,000 parasite proteins across five different species by extensive subcellular fractionation of erythrocytes infected by Plasmodium falciparum, Plasmodium knowlesi, Plasmodium yoelii, Plasmodium berghei, and Plasmodium chabaudi. This comparison of the pan-species spatial proteomes and their expression patterns indicates increasing species-specific proteins associated with the more external compartments, supporting host adaptations and post-transcriptional regulation. The spatial proteome offers comprehensive insight into the different human, simian, and rodent Plasmodium species, establishing a powerful resource for understanding species-specific host adaptation processes in the parasite.


Asunto(s)
Malaria , Proteómica , Humanos , Malaria/parasitología , Proteoma/metabolismo , Plasmodium berghei/metabolismo , Eritrocitos/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA