Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
ACS Infect Dis ; 10(4): 1286-1297, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38556981

RESUMEN

Malaria is caused by parasites of the Plasmodium genus and remains one of the most pressing human health problems. The spread of parasites resistant to or partially resistant to single or multiple drugs, including frontline antimalarial artemisinin and its derivatives, poses a serious threat to current and future malaria control efforts. In vitro drug assays are important for identifying new antimalarial compounds and monitoring drug resistance. Due to its robustness and ease of use, the [3H]-hypoxanthine incorporation assay is still considered a gold standard and is widely applied, despite limited sensitivity and the dependence on radioactive material. Here, we present a first-of-its-kind chemiluminescence-based antimalarial drug screening assay. The effect of compounds on P. falciparum is monitored by using a dioxetane-based substrate (AquaSpark ß-D-galactoside) that emits high-intensity luminescence upon removal of a protective group (ß-D-galactoside) by a transgenic ß-galactosidase reporter enzyme. This biosensor enables highly sensitive, robust, and cost-effective detection of asexual, intraerythrocytic P. falciparum parasites without the need for parasite enrichment, washing, or purification steps. We are convinced that the ultralow detection limit of less than 100 parasites of the presented biosensor system will become instrumental in malaria research, including but not limited to drug screening.


Asunto(s)
Antimaláricos , Antagonistas del Ácido Fólico , Malaria Falciparum , Malaria , Humanos , Antimaláricos/farmacología , Plasmodium falciparum , Malaria/tratamiento farmacológico , Malaria Falciparum/parasitología , Antagonistas del Ácido Fólico/farmacología , Galactósidos/farmacología , Galactósidos/uso terapéutico
2.
Cell Rep ; 43(4): 114012, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38573856

RESUMEN

Plasmodium falciparum is a human-adapted apicomplexan parasite that causes the most dangerous form of malaria. P. falciparum cysteine-rich protective antigen (PfCyRPA) is an invasion complex protein essential for erythrocyte invasion. The precise role of PfCyRPA in this process has not been resolved. Here, we show that PfCyRPA is a lectin targeting glycans terminating with α2-6-linked N-acetylneuraminic acid (Neu5Ac). PfCyRPA has a >50-fold binding preference for human, α2-6-linked Neu5Ac over non-human, α2-6-linked N-glycolylneuraminic acid. PfCyRPA lectin sites were predicted by molecular modeling and validated by mutagenesis studies. Transgenic parasite lines expressing endogenous PfCyRPA with single amino acid exchange mutants indicated that the lectin activity of PfCyRPA has an important role in parasite invasion. Blocking PfCyRPA lectin activity with small molecules or with lectin-site-specific monoclonal antibodies can inhibit blood-stage parasite multiplication. Therefore, targeting PfCyRPA lectin activity with drugs, immunotherapy, or a vaccine-primed immune response is a promising strategy to prevent and treat malaria.


Asunto(s)
Eritrocitos , Plasmodium falciparum , Polisacáridos , Proteínas Protozoarias , Humanos , Antígenos de Protozoos/metabolismo , Antígenos de Protozoos/inmunología , Antígenos de Protozoos/genética , Eritrocitos/parasitología , Eritrocitos/metabolismo , Lectinas/metabolismo , Lectinas/genética , Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Polisacáridos/metabolismo , Unión Proteica , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética
3.
Elife ; 122023 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-37934560

RESUMEN

Plasmodium falciparum accounts for the majority of over 600,000 malaria-associated deaths annually. Parasites resistant to nearly all antimalarials have emerged and the need for drugs with alternative modes of action is thus undoubted. The FK506-binding protein PfFKBP35 has gained attention as a promising drug target due to its high affinity to the macrolide compound FK506 (tacrolimus). Whilst there is considerable interest in targeting PfFKBP35 with small molecules, a genetic validation of this factor as a drug target is missing and its function in parasite biology remains elusive. Here, we show that limiting PfFKBP35 levels are lethal to P. falciparum and result in a delayed death-like phenotype that is characterized by defective ribosome homeostasis and stalled protein synthesis. Our data furthermore suggest that FK506, unlike the action of this drug in model organisms, exerts its antiproliferative activity in a PfFKBP35-independent manner and, using cellular thermal shift assays, we identify putative FK506-targets beyond PfFKBP35. In addition to revealing first insights into the function of PfFKBP35, our results show that FKBP-binding drugs can adopt non-canonical modes of action - with major implications for the development of FK506-derived molecules active against Plasmodium parasites and other eukaryotic pathogens.


Asunto(s)
Antimaláricos , Malaria Falciparum , Humanos , Tacrolimus , Antibacterianos , Sistemas de Liberación de Medicamentos , Homeostasis , Proteínas de Unión a Tacrolimus
4.
Brief Bioinform ; 24(4)2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37406192

RESUMEN

Recent advances in long read technologies not only enable large consortia to aim to sequence all eukaryotes on Earth, but they also allow individual laboratories to sequence their species of interest with relatively low investment. Long read technologies embody the promise of overcoming scaffolding problems associated with repeats and low complexity sequences, but the number of contigs often far exceeds the number of chromosomes and they may contain many insertion and deletion errors around homopolymer tracts. To overcome these issues, we have implemented the ILRA pipeline to correct long read-based assemblies. Contigs are first reordered, renamed, merged, circularized, or filtered if erroneous or contaminated. Illumina short reads are used subsequently to correct homopolymer errors. We successfully tested our approach by improving the genome sequences of Homo sapiens, Trypanosoma brucei, and Leptosphaeria spp., and by generating four novel Plasmodium falciparum assemblies from field samples. We found that correcting homopolymer tracts reduced the number of genes incorrectly annotated as pseudogenes, but an iterative approach seems to be required to correct more sequencing errors. In summary, we describe and benchmark the performance of our new tool, which improved the quality of novel long read assemblies up to 1 Gbp. The pipeline is available at GitHub: https://github.com/ThomasDOtto/ILRA.


Asunto(s)
Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Análisis de Secuencia de ADN , Seudogenes , Cromosomas
5.
Front Cell Infect Microbiol ; 12: 802341, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35223540

RESUMEN

Malaria parasites rely on specialized stages, called gametocytes, to ensure human-to-human transmission. The formation of these sexual precursor cells is initiated by commitment of blood stage parasites to the sexual differentiation pathway. Plasmodium falciparum, the most virulent of six parasite species infecting humans, employs nutrient sensing to control the rate at which sexual commitment is initiated, and the presence of stress-inducing factors, including antimalarial drugs, has been linked to increased gametocyte production in vitro and in vivo. These observations suggest that therapeutic interventions may promote gametocytogenesis and malaria transmission. Here, we engineered a P. falciparum reporter line to quantify sexual commitment rates after exposure to antimalarials and other pharmaceuticals commonly prescribed in malaria-endemic regions. Our data reveal that some of the tested drugs indeed have the capacity to elevate sexual commitment rates in vitro. Importantly, however, these effects are only observed at drug concentrations that inhibit parasite survival and only rarely result in a net increase of gametocyte production. Using a drug-resistant parasite reporter line, we further show that the gametocytogenesis-promoting effect of drugs is linked to general stress responses rather than to compound-specific activities. Altogether, we did not observe evidence for mechanistic links between the regulation of sexual commitment and the activity of commonly used pharmaceuticals in vitro. Our data hence does not support scenarios in which currently applied therapeutic interventions would promote the spread of drug-resistant parasites or malaria transmission in general.


Asunto(s)
Antimaláricos , Malaria Falciparum , Malaria , Parásitos , Animales , Antimaláricos/farmacología , Humanos , Malaria/parasitología , Malaria Falciparum/parasitología , Preparaciones Farmacéuticas , Plasmodium falciparum
6.
PLoS Biol ; 19(12): e3001483, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34879056

RESUMEN

Cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) signalling is essential for the proliferation of Plasmodium falciparum malaria blood stage parasites. The mechanisms regulating the activity of the catalytic subunit PfPKAc, however, are only partially understood, and PfPKAc function has not been investigated in gametocytes, the sexual blood stage forms that are essential for malaria transmission. By studying a conditional PfPKAc knockdown (cKD) mutant, we confirm the essential role for PfPKAc in erythrocyte invasion by merozoites and show that PfPKAc is involved in regulating gametocyte deformability. We furthermore demonstrate that overexpression of PfPKAc is lethal and kills parasites at the early phase of schizogony. Strikingly, whole genome sequencing (WGS) of parasite mutants selected to tolerate increased PfPKAc expression levels identified missense mutations exclusively in the gene encoding the parasite orthologue of 3-phosphoinositide-dependent protein kinase-1 (PfPDK1). Using targeted mutagenesis, we demonstrate that PfPDK1 is required to activate PfPKAc and that T189 in the PfPKAc activation loop is the crucial target residue in this process. In summary, our results corroborate the importance of tight regulation of PfPKA signalling for parasite survival and imply that PfPDK1 acts as a crucial upstream regulator in this pathway and potential new drug target.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Animales , Dominio Catalítico , Línea Celular , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/genética , Eritrocitos/parasitología , Humanos , Malaria , Malaria Falciparum/parasitología , Merozoítos , Parásitos/metabolismo , Proteínas Protozoarias/metabolismo
7.
Nat Commun ; 12(1): 4806, 2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34376675

RESUMEN

The malaria parasite Plasmodium falciparum replicates inside erythrocytes in the blood of infected humans. During each replication cycle, a small proportion of parasites commits to sexual development and differentiates into gametocytes, which are essential for parasite transmission via the mosquito vector. Detailed molecular investigation of gametocyte biology and transmission has been hampered by difficulties in generating large numbers of these highly specialised cells. Here, we engineer P. falciparum NF54 inducible gametocyte producer (iGP) lines for the routine mass production of synchronous gametocytes via conditional overexpression of the sexual commitment factor GDV1. NF54/iGP lines consistently achieve sexual commitment rates of 75% and produce viable gametocytes that are transmissible by mosquitoes. We also demonstrate that further genetic engineering of NF54/iGP parasites is a valuable tool for the targeted exploration of gametocyte biology. In summary, we believe the iGP approach developed here will greatly expedite basic and applied malaria transmission stage research.


Asunto(s)
Sistemas CRISPR-Cas , Malaria Falciparum/sangre , Plasmodium falciparum/genética , Esporas Protozoarias/genética , Animales , Anopheles/parasitología , Células Cultivadas , Eritrocitos/parasitología , Hepatocitos/citología , Hepatocitos/parasitología , Interacciones Huésped-Parásitos , Humanos , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Microscopía Fluorescente , Mosquitos Vectores/parasitología , Plasmodium falciparum/fisiología , Esporas Protozoarias/fisiología , Esporozoítos/genética , Esporozoítos/fisiología
8.
Commun Biol ; 4(1): 336, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712726

RESUMEN

Casein kinase 2 (CK2) is a pleiotropic kinase phosphorylating substrates in different cellular compartments in eukaryotes. In the malaria parasite Plasmodium falciparum, PfCK2 is vital for asexual proliferation of blood-stage parasites. Here, we applied CRISPR/Cas9-based gene editing to investigate the function of the PfCK2α catalytic subunit in gametocytes, the sexual forms of the parasite that are essential for malaria transmission. We show that PfCK2α localizes to the nucleus and cytoplasm in asexual and sexual parasites alike. Conditional knockdown of PfCK2α expression prevented the transition of stage IV into transmission-competent stage V gametocytes, whereas the conditional knockout of pfck2a completely blocked gametocyte maturation already at an earlier stage of sexual differentiation. In summary, our results demonstrate that PfCK2α is not only essential for asexual but also sexual development of P. falciparum blood-stage parasites and encourage studies exploring PfCK2α as a potential target for dual-active antimalarial drugs.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Eritrocitos/parasitología , Gametogénesis , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Antimaláricos/farmacología , Sistemas CRISPR-Cas , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/genética , Dominio Catalítico , Edición Génica , Humanos , Estadios del Ciclo de Vida , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Protozoarias/genética , Reproducción Asexuada
9.
mSphere ; 6(1)2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33536327

RESUMEN

The human malaria parasite Plasmodium falciparum encodes a single ortholog of heterochromatin protein 1 (PfHP1) that plays a crucial role in the epigenetic regulation of various survival-related processes. PfHP1 is essential for parasite proliferation and the heritable silencing of genes linked to antigenic variation, host cell invasion, and sexual conversion. Here, we employed CRISPR/Cas9-mediated genome editing combined with the DiCre/loxP system to investigate how the PfHP1 chromodomain (CD), hinge domain, and chromoshadow domain (CSD) contribute to overall PfHP1 function. We show that the 76 C-terminal residues are responsible for targeting PfHP1 to the nucleus. Furthermore, we reveal that each of the three functional domains of PfHP1 are required for heterochromatin formation, gene silencing, and mitotic parasite proliferation. Finally, we discovered that the hinge domain and CSD of HP1 are functionally conserved between P. falciparum and P. berghei, a related malaria parasite infecting rodents. In summary, our study provides new insights into PfHP1 function and offers a tool for further studies on epigenetic regulation and life cycle decision in malaria parasites.IMPORTANCE Malaria is caused by unicellular Plasmodium species parasites that repeatedly invade and replicate inside red blood cells. Some blood-stage parasites exit the cell cycle and differentiate into gametocytes that are essential for malaria transmission via the mosquito vector. Epigenetic control mechanisms allow the parasites to alter the expression of surface antigens and to balance the switch between parasite multiplication and gametocyte production. These processes are crucial to establish chronic infection and optimize parasite transmission. Here, we performed a mutational analysis of heterochromatin protein 1 (HP1) in P. falciparum We demonstrate that all three domains of this protein are indispensable for the proper function of HP1 in parasite multiplication, heterochromatin formation, and gene silencing. Moreover, expression of chimeric proteins revealed the functional conservation of HP1 proteins between different Plasmodium species. These results provide new insight into the function and evolution of HP1 as an essential epigenetic regulator of parasite survival.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Silenciador del Gen , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Variación Antigénica , Sistemas CRISPR-Cas , Línea Celular , Homólogo de la Proteína Chromobox 5 , Eritrocitos/parasitología , Regulación de la Expresión Génica , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo
10.
PLoS One ; 15(9): e0238134, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32936796

RESUMEN

Malaria is a life-threatening disease, caused by Apicomplexan parasites of the Plasmodium genus. The Anopheles mosquito is necessary for the sexual replication of these parasites and for their transmission to vertebrate hosts, including humans. Imaging of the parasite within the insect vector has been attempted using multiple microscopy methods, most of which are hampered by the presence of the light scattering opaque cuticle of the mosquito. So far, most imaging of the Plasmodium mosquito stages depended on either sectioning or surgical dissection of important anatomical sites, such as the midgut and the salivary glands. Optical projection tomography (OPT) and light sheet fluorescence microscopy (LSFM) enable imaging fields of view in the centimeter scale whilst providing micrometer resolution. In this paper, we compare different optical clearing protocols and present reconstructions of the whole body of Plasmodium-infected, optically cleared Anopheles stephensi mosquitoes and their midguts. The 3D-reconstructions from OPT imaging show detailed features of the mosquito anatomy and enable overall localization of parasites in midguts. Additionally, LSFM imaging of mosquito midguts shows detailed distribution of oocysts in extracted midguts. This work was submitted as a pre-print to bioRxiv, available at https://www.biorxiv.org/content/10.1101/682054v2.


Asunto(s)
Anopheles/fisiología , Imagenología Tridimensional , Insectos Vectores/fisiología , Microscopía Fluorescente , Plasmodium/fisiología , Tomografía Óptica , Animales , Anopheles/parasitología
11.
Trends Parasitol ; 36(2): 85-87, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31883707

RESUMEN

A recent report by Jennison et al. reveals an important role for plasmepsin V (PMV), an aspartyl protease, in the development of malaria transmission stages. The authors showed that PMV activity is critical for protein export in these stages and that specific PMV inhibitors block parasite transmission to mosquitoes.


Asunto(s)
Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/metabolismo , Malaria/parasitología , Malaria/transmisión , Plasmodium/enzimología , Animales , Antimaláricos/farmacología , Carbamatos/farmacología , Humanos , Estadios del Ciclo de Vida/efectos de los fármacos , Estadios del Ciclo de Vida/fisiología , Malaria/prevención & control , Oligopéptidos/farmacología , Plasmodium/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos
12.
Sci Rep ; 9(1): 16720, 2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31723180

RESUMEN

Previous studies in model eukaryotes have demonstrated that phosphorylation of heterochromatin protein 1 (HP1) is important for dynamically regulating its various functions. However, in the malaria parasite Plasmodium falciparum both the function of HP1 phosphorylation and the identity of the protein kinases targeting HP1 are still elusive. In order to functionally analyze phosphorylation of P. falciparum HP1 (PfHP1), we first mapped PfHP1 phosphorylation sites by liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis of native PfHP1, which identified motifs from which potential kinases could be predicted; in particular, several phosphorylated residues were embedded in motifs rich in acidic residues, reminiscent of targets for P. falciparum casein kinase 2 (PfCK2). Secondly, we tested recombinant PfCK2 and a number of additional protein kinases for their ability to phosphorylate PfHP1 in in vitro kinase assays. These experiments validated our prediction that PfHP1 acts as a substrate for PfCK2. Furthermore, LC-MS/MS analysis showed that PfCK2 phosphorylates three clustered serine residues in an acidic motif within the central hinge region of PfHP1. To study the role of PfHP1 phosphorylation in live parasites we used CRISPR/Cas9-mediated genome editing to generate a number of conditional PfHP1 phosphomutants based on the DiCre/LoxP system. Our studies revealed that neither PfCK2-dependent phosphorylation of PfHP1, nor phosphorylation of the hinge domain in general, affect PfHP1's ability to localize to heterochromatin, and that PfHP1 phosphorylation in this region is dispensable for the proliferation of P. falciparum blood stage parasites.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/metabolismo , Malaria Falciparum/parasitología , Plasmodium falciparum/aislamiento & purificación , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Secuencia de Aminoácidos , Quinasa de la Caseína II/genética , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Humanos , Malaria Falciparum/metabolismo , Mutación , Fosforilación , Proteínas Protozoarias/genética
13.
Science ; 365(6456)2019 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-31467193

RESUMEN

The requirement for next-generation antimalarials to be both curative and transmission-blocking necessitates the identification of previously undiscovered druggable molecular pathways. We identified a selective inhibitor of the Plasmodium falciparum protein kinase PfCLK3, which we used in combination with chemogenetics to validate PfCLK3 as a drug target acting at multiple parasite life stages. Consistent with a role for PfCLK3 in RNA splicing, inhibition resulted in the down-regulation of more than 400 essential parasite genes. Inhibition of PfCLK3 mediated rapid killing of asexual liver- and blood-stage P. falciparum and blockade of gametocyte development, thereby preventing transmission, and also showed parasiticidal activity against P. berghei and P. knowlesi Hence, our data establish PfCLK3 as a target for drugs, with the potential to offer a cure-to be prophylactic and transmission blocking in malaria.


Asunto(s)
Antimaláricos/farmacología , Terapia Molecular Dirigida , Plasmodium falciparum/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Protozoarias/antagonistas & inhibidores , Animales , Antimaláricos/química , Antimaláricos/aislamiento & purificación , Antimaláricos/uso terapéutico , Gametogénesis/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Ratones , Ratones Endogámicos BALB C , Plasmodium falciparum/enzimología , Plasmodium falciparum/genética , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteínas Protozoarias/genética , Empalme del ARN/genética , Bibliotecas de Moléculas Pequeñas/farmacología
14.
Cell Microbiol ; 21(5): e13023, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30825872

RESUMEN

Intravital microscopy allows the visualisation of how pathogens interact with host cells and tissues in living animals in real time. This method has enabled key advances in our understanding of host-parasite interactions under physiological conditions. A combination of genetics, microscopy techniques, and image analysis have recently facilitated the understanding of biological phenomena in living animals at cellular and subcellular resolution. In this review, we summarise findings achieved by intravital microscopy of the skin and adipose tissues upon infection with various parasites, and we present a view into possible future applications of this method.


Asunto(s)
Tejido Adiposo/diagnóstico por imagen , Tejido Adiposo/parasitología , Interacciones Huésped-Parásitos , Microscopía Intravital/métodos , Piel/diagnóstico por imagen , Piel/parasitología , Tejido Adiposo/citología , Tejido Adiposo/patología , Animales , Microscopía Intravital/tendencias , Leishmania/metabolismo , Leishmania/patogenicidad , Plasmodium/metabolismo , Plasmodium/patogenicidad , Schistosoma/metabolismo , Schistosoma/patogenicidad , Piel/citología , Piel/patología , Trypanosoma/metabolismo , Trypanosoma/patogenicidad
15.
Wellcome Open Res ; 3: 70, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30320226

RESUMEN

Background: Malaria parasites go through major transitions during their complex life cycle, yet the underlying differentiation pathways remain obscure. Here we apply single cell transcriptomics to unravel the program inducing sexual differentiation in Plasmodium falciparum. Parasites have to make this essential life-cycle decision in preparation for human-to-mosquito transmission. Methods: By combining transcriptional profiling with quantitative imaging and genetics, we defined a transcriptional signature in sexually committed cells. Results: We found this transcriptional signature to be distinct from general changes in parasite metabolism that can be observed in response to commitment-inducing conditions. Conclusions: This proof-of-concept study provides a template to capture transcriptional diversity in parasite populations containing complex mixtures of different life-cycle stages and developmental programs, with important implications for our understanding of parasite biology and the ongoing malaria elimination campaign.

16.
Sci Adv ; 4(5): eaat3775, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29806032

RESUMEN

Transmission of Plasmodium parasites to the mosquito requires the formation and development of gametocytes. Studies in infected humans have shown that only the most mature forms of Plasmodium falciparum gametocytes are present in circulation, whereas immature forms accumulate in the hematopoietic environment of the bone marrow. We used the rodent model Plasmodium berghei to study gametocyte behavior through time under physiological conditions. Intravital microscopy demonstrated preferential homing of early gametocyte forms across the intact vascular barrier of the bone marrow and the spleen early during infection and subsequent development in the extravascular environment. During the acute phase of infection, we observed vascular leakage resulting in further parasite accumulation in this environment. Mature gametocytes showed high deformability and were found entering and exiting the intact vascular barrier. We suggest that extravascular gametocyte localization and mobility are essential for gametocytogenesis and transmission of Plasmodium to the mosquito.


Asunto(s)
Médula Ósea/parasitología , Malaria/patología , Malaria/parasitología , Plasmodium/fisiología , Migración Transendotelial y Transepitelial , Animales , Modelos Animales de Enfermedad , Interacciones Huésped-Parásitos , Humanos , Ratones , Imagen Molecular , Sistema Mononuclear Fagocítico/parasitología
17.
Science ; 359(6381): 1259-1263, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29590075

RESUMEN

Malaria is caused by Plasmodium parasites that proliferate in the bloodstream. During each replication cycle, some parasites differentiate into gametocytes, the only forms able to infect the mosquito vector and transmit malaria. Sexual commitment is triggered by activation of AP2-G, the master transcriptional regulator of gametocytogenesis. Heterochromatin protein 1 (HP1)-dependent silencing of ap2-g prevents sexual conversion in proliferating parasites. In this study, we identified Plasmodium falciparum gametocyte development 1 (GDV1) as an upstream activator of sexual commitment. We found that GDV1 targeted heterochromatin and triggered HP1 eviction, thus derepressing ap2-g Expression of GDV1 was responsive to environmental triggers of sexual conversion and controlled via a gdv1 antisense RNA. Hence, GDV1 appears to act as an effector protein that induces sexual differentiation by antagonizing HP1-dependent gene silencing.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Gametogénesis/genética , Silenciador del Gen , Malaria Falciparum/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Diferenciación Sexual/genética , Animales , Homólogo de la Proteína Chromobox 5 , Plasmodium falciparum/genética
18.
Cell Host Microbe ; 23(3): 407-420.e8, 2018 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-29503181

RESUMEN

Heterochromatin-dependent gene silencing is central to the adaptation and survival of Plasmodium falciparum malaria parasites, allowing clonally variant gene expression during blood infection in humans. By assessing genome-wide heterochromatin protein 1 (HP1) occupancy, we present a comprehensive analysis of heterochromatin landscapes across different Plasmodium species, strains, and life cycle stages. Common targets of epigenetic silencing include fast-evolving multi-gene families encoding surface antigens and a small set of conserved HP1-associated genes with regulatory potential. Many P. falciparum heterochromatic genes are marked in a strain-specific manner, increasing the parasite's adaptive capacity. Whereas heterochromatin is strictly maintained during mitotic proliferation of asexual blood stage parasites, substantial heterochromatin reorganization occurs in differentiating gametocytes and appears crucial for the activation of key gametocyte-specific genes and adaptation of erythrocyte remodeling machinery. Collectively, these findings provide a catalog of heterochromatic genes and reveal conserved and specialized features of epigenetic control across the genus Plasmodium.


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Epigénesis Genética/genética , Epigenómica , Perfilación de la Expresión Génica , Heterocromatina/genética , Plasmodium/genética , Plasmodium/fisiología , Adaptación Fisiológica/genética , Adaptación Fisiológica/fisiología , Animales , Variación Antigénica/genética , Antígenos de Protozoos/genética , Proliferación Celular , Homólogo de la Proteína Chromobox 5 , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Silenciador del Gen , Interacciones Huésped-Parásitos/genética , Interacciones Huésped-Parásitos/fisiología , Humanos , Estadios del Ciclo de Vida/genética , Estadios del Ciclo de Vida/fisiología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos BALB C , Parásitos/genética , Filogenia , Plasmodium/clasificación , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Diferenciación Sexual
19.
Cell ; 171(7): 1532-1544.e15, 2017 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-29129376

RESUMEN

Transmission represents a population bottleneck in the Plasmodium life cycle and a key intervention target of ongoing efforts to eradicate malaria. Sexual differentiation is essential for this process, as only sexual parasites, called gametocytes, are infective to the mosquito vector. Gametocyte production rates vary depending on environmental conditions, but external stimuli remain obscure. Here, we show that the host-derived lipid lysophosphatidylcholine (LysoPC) controls P. falciparum cell fate by repressing parasite sexual differentiation. We demonstrate that exogenous LysoPC drives biosynthesis of the essential membrane component phosphatidylcholine. LysoPC restriction induces a compensatory response, linking parasite metabolism to the activation of sexual-stage-specific transcription and gametocyte formation. Our results reveal that malaria parasites can sense and process host-derived physiological signals to regulate differentiation. These data close a critical knowledge gap in parasite biology and introduce a major component of the sexual differentiation pathway in Plasmodium that may provide new approaches for blocking malaria transmission.


Asunto(s)
Lisofosfatidilcolinas/metabolismo , Malaria/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/metabolismo , Animales , Femenino , Humanos , Malaria/inmunología , Redes y Vías Metabólicas , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/fisiología , Reproducción
20.
Nat Commun ; 7: 12727, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27721445

RESUMEN

Malaria remains one of the greatest public health challenges worldwide, particularly in sub-Saharan Africa. The clinical outcome of individuals infected with Plasmodium falciparum parasites depends on many factors including host systemic inflammatory responses, parasite sequestration in tissues and vascular dysfunction. Production of pro-inflammatory cytokines and chemokines promotes endothelial activation as well as recruitment and infiltration of inflammatory cells, which in turn triggers further endothelial cell activation and parasite sequestration. Inflammatory responses are triggered in part by bioactive parasite products such as hemozoin and infected red blood cell-derived extracellular vesicles (iRBC-derived EVs). Here we demonstrate that such EVs contain functional miRNA-Argonaute 2 complexes that are derived from the host RBC. Moreover, we show that EVs are efficiently internalized by endothelial cells, where the miRNA-Argonaute 2 complexes modulate target gene expression and barrier properties. Altogether, these findings provide a mechanistic link between EVs and vascular dysfunction during malaria infection.


Asunto(s)
Proteínas Argonautas/metabolismo , Vasos Sanguíneos/metabolismo , Eritrocitos/parasitología , Vesículas Extracelulares/metabolismo , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , MicroARNs/metabolismo , Encéfalo/irrigación sanguínea , Línea Celular , Endocitosis , Células Endoteliales/metabolismo , Eritrocitos/ultraestructura , Vesículas Extracelulares/ultraestructura , Regulación de la Expresión Génica , Silenciador del Gen , Humanos , MicroARNs/genética , Microvasos/citología , Complejo Silenciador Inducido por ARN/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA