Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 179(5): 1112-1128.e26, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31730853

RESUMEN

Plasmodium gene functions in mosquito and liver stages remain poorly characterized due to limitations in the throughput of phenotyping at these stages. To fill this gap, we followed more than 1,300 barcoded P. berghei mutants through the life cycle. We discover 461 genes required for efficient parasite transmission to mosquitoes through the liver stage and back into the bloodstream of mice. We analyze the screen in the context of genomic, transcriptomic, and metabolomic data by building a thermodynamic model of P. berghei liver-stage metabolism, which shows a major reprogramming of parasite metabolism to achieve rapid growth in the liver. We identify seven metabolic subsystems that become essential at the liver stages compared with asexual blood stages: type II fatty acid synthesis and elongation (FAE), tricarboxylic acid, amino sugar, heme, lipoate, and shikimate metabolism. Selected predictions from the model are individually validated in single mutants to provide future targets for drug development.


Asunto(s)
Genoma de Protozoos , Estadios del Ciclo de Vida/genética , Hígado/metabolismo , Hígado/parasitología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/genética , Alelos , Amino Azúcares/biosíntesis , Animales , Culicidae/parasitología , Eritrocitos/parasitología , Ácido Graso Sintasas/metabolismo , Ácidos Grasos/metabolismo , Técnicas de Inactivación de Genes , Genotipo , Modelos Biológicos , Mutación/genética , Parásitos/genética , Parásitos/crecimiento & desarrollo , Fenotipo , Plasmodium berghei/metabolismo , Ploidias , Reproducción
2.
Proc Natl Acad Sci U S A ; 119(35): e2209729119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35994647

RESUMEN

Glutaminyl cyclase (QC) modifies N-terminal glutamine or glutamic acid residues of target proteins into cyclic pyroglutamic acid (pGlu). Here, we report the biochemical and functional analysis of Plasmodium QC. We show that sporozoites of QC-null mutants of rodent and human malaria parasites are recognized by the mosquito immune system and melanized when they reach the hemocoel. Detailed analyses of rodent malaria QC-null mutants showed that sporozoite numbers in salivary glands are reduced in mosquitoes infected with QC-null or QC catalytically dead mutants. This phenotype can be rescued by genetic complementation or by disrupting mosquito melanization or phagocytosis by hemocytes. Mutation of a single QC-target glutamine of the major sporozoite surface protein (circumsporozoite protein; CSP) of the rodent parasite Plasmodium berghei also results in melanization of sporozoites. These findings indicate that QC-mediated posttranslational modification of surface proteins underlies evasion of killing of sporozoites by the mosquito immune system.


Asunto(s)
Aminoaciltransferasas , Culicidae , Malaria , Procesamiento Proteico-Postraduccional , Esporozoítos , Aminoaciltransferasas/inmunología , Animales , Culicidae/inmunología , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Malaria/genética , Malaria/inmunología , Malaria/parasitología , Plasmodium berghei/genética , Plasmodium berghei/inmunología , Procesamiento Proteico-Postraduccional/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología
3.
Malar J ; 23(1): 111, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38641838

RESUMEN

BACKGROUND: Sporozoites (SPZ), the infective form of Plasmodium falciparum malaria, can be inoculated into the human host skin by Anopheline mosquitoes. These SPZ migrate at approximately 1 µm/s to find a blood vessel and travel to the liver where they infect hepatocytes and multiply. In the skin they are still low in number (50-100 SPZ) and vulnerable to immune attack by antibodies and skin macrophages. This is why whole SPZ and SPZ proteins are used as the basis for most malaria vaccines currently deployed and undergoing late clinical testing. Mosquitoes typically inoculate SPZ into a human host between 14 and 25 days after their previous infective blood meal. However, it is unknown whether residing time within the mosquito affects SPZ condition, infectivity or immunogenicity. This study aimed to unravel how the age of P. falciparum SPZ in salivary glands (14, 17, or 20 days post blood meal) affects their infectivity and the ensuing immune responses. METHODS: SPZ numbers, viability by live/dead staining, motility using dedicated sporozoite motility orienting and organizing tool software (SMOOT), and infectivity of HC-04.j7 liver cells at 14, 17 and 20 days after mosquito feeding have been investigated. In vitro co-culture assays with SPZ stimulated monocyte-derived macrophages (MoMɸ) and CD8+ T-cells, analysed by flow cytometry, were used to investigate immune responses. RESULTS: SPZ age did not result in different SPZ numbers or viability. However, a markedly different motility pattern, whereby motility decreased from 89% at day 14 to 80% at day 17 and 71% at day 20 was observed (p ≤ 0.0001). Similarly, infectivity of day 20 SPZ dropped to ~ 50% compared with day 14 SPZ (p = 0.004). MoMɸ were better able to take up day 14 SPZ than day 20 SPZ (from 7.6% to 4.1%, p = 0.03) and displayed an increased expression of pro-inflammatory CD80, IL-6 (p = 0.005), regulatory markers PDL1 (p = 0.02), IL-10 (p = 0.009) and cytokines upon phagocytosis of younger SPZ. Interestingly, co-culture of these cells with CD8+ T-cells revealed a decreased expression of activation marker CD137 and cytokine IFNγ compared to their day 20 counterparts. These findings suggest that older (day 17-20) P. falciparum SPZ are less infectious and have decreased immune regulatory potential. CONCLUSION: Overall, this data is a first step in enhancing the understanding of how mosquito residing time affects P. falciparum SPZ and could impact the understanding of the P. falciparum infectious reservoir and the potency of whole SPZ vaccines.


Asunto(s)
Culicidae , Vacunas contra la Malaria , Malaria Falciparum , Animales , Humanos , Esporozoítos , Linfocitos T CD8-positivos , Envejecimiento , Plasmodium falciparum
4.
PLoS Pathog ; 17(11): e1010114, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34843584

RESUMEN

Malaria is a hazardous disease caused by Plasmodium parasites and often results in lethal complications, including malaria-associated acute respiratory distress syndrome (MA-ARDS). Parasite sequestration in the microvasculature is often observed, but its role in malaria pathogenesis and complications is still incompletely understood. We used skeleton binding protein-1 (SBP-1) KO parasites to study the role of sequestration in experimental MA-ARDS. The sequestration-deficiency of these SBP-1 KO parasites was confirmed with bioluminescence imaging and by measuring parasite accumulation in the lungs with RT-qPCR. The SBP-1 KO parasites induced similar lung pathology in the early stage of experimental MA-ARDS compared to wildtype (WT) parasites. Strikingly, the lung pathology resolved subsequently in more than 60% of the SBP-1 KO infected mice, resulting in prolonged survival despite the continuous presence of the parasite. This spontaneous disease resolution was associated with decreased inflammatory cytokine expression measured by RT-qPCR and lower expression of cytotoxic markers in pathogenic CD8+ T cells in the lungs of SBP-1 KO infected mice. These data suggest that SBP-1-mediated parasite sequestration and subsequent high parasite load are not essential for the development of experimental MA-ARDS but inhibit the resolution of the disease.


Asunto(s)
Pulmón/parasitología , Malaria/complicaciones , Proteínas de la Membrana/deficiencia , Plasmodium berghei/patogenicidad , Proteínas Protozoarias/metabolismo , Síndrome de Dificultad Respiratoria/prevención & control , Animales , Progresión de la Enfermedad , Femenino , Pulmón/metabolismo , Pulmón/patología , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Protozoarias/genética , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/parasitología , Síndrome de Dificultad Respiratoria/patología
5.
PLoS Pathog ; 16(9): e1008799, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32898164

RESUMEN

Professional antigen-presenting cells (APCs), like macrophages (Mϕs) and dendritic cells (DCs), are central players in the induction of natural and vaccine-induced immunity to malaria, yet very little is known about the interaction of SPZ with human APCs. Intradermal delivery of whole-sporozoite vaccines reduces their effectivity, possibly due to dermal immunoregulatory effects. Therefore, understanding these interactions could prove pivotal to malaria vaccination. We investigated human APC responses to recombinant circumsporozoite protein (recCSP), SPZ and anti-CSP opsonized SPZ both in monocyte derived MoDCs and MoMϕs. Both MoDCs and MoMϕs readily took up recCSP but did not change phenotype or function upon doing so. SPZ are preferentially phagocytosed by MoMϕs instead of DCs and phagocytosis greatly increased after opsonization. Subsequently MoMϕs show increased surface marker expression of activation markers as well as tolerogenic markers such as Programmed Death-Ligand 1 (PD-L1). Additionally they show reduced motility, produce interleukin 10 and suppressed interferon gamma (IFNγ) production by antigen specific CD8+ T cells. Importantly, we investigated phenotypic responses to SPZ in primary dermal APCs isolated from human skin explants, which respond similarly to their monocyte-derived counterparts. These findings are a first step in enhancing our understanding of pre-erythrocytic natural immunity and the pitfalls of intradermal vaccination-induced immunity.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Macrófagos/inmunología , Malaria/inmunología , Plasmodium berghei/inmunología , Proteínas Protozoarias/inmunología , Piel/inmunología , Esporozoítos/inmunología , Animales , Células Cultivadas , Femenino , Humanos , Macrófagos/parasitología , Malaria/parasitología , Ratones , Piel/parasitología
6.
FASEB J ; 35(12): e21997, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34719814

RESUMEN

The deadliest complication of infection by Plasmodium parasites, cerebral malaria, accounts for the majority of malarial fatalities. Although our understanding of the cellular and molecular mechanisms underlying the pathology remains incomplete, recent studies support the contribution of systemic and neuroinflammation as the cause of cerebral edema and blood-brain barrier (BBB) dysfunction. All Plasmodium species encode an orthologue of the innate cytokine, Macrophage Migration Inhibitory Factor (MIF), which functions in mammalian biology to regulate innate responses. Plasmodium MIF (PMIF) similarly signals through the host MIF receptor CD74, leading to an enhanced inflammatory response. We investigated the PMIF-CD74 interaction in the onset of experimental cerebral malaria (ECM) and liver stage Plasmodium development by using a combination of CD74 deficient (Cd74-/- ) hosts and PMIF deficient parasites. Cd74-/- mice were found to be protected from ECM and the protection was associated with the inability of brain microvessels to present parasite antigen to sequestered and pathogenic Plasmodium-specific CD8+ T cells. Infection of WT hosts with PMIF-deficient sporozoites or infection of Cd74-/- hosts with WT sporozoites impacted the survival of infected hepatocytes and subsequently reduced blood-stage associated inflammation, contributing to protection from ECM. We recapitulated these finding with a novel pharmacologic PMIF-selective antagonist that reduced PMIF/CD74 signaling and fully protected mice from ECM. These findings reveal a conserved mechanism for Plasmodium usurpation of host CD74 signaling and suggest a tractable approach for new pharmacologic intervention.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/química , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase II/química , Inflamación/prevención & control , Hígado/patología , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Malaria Cerebral/prevención & control , Plasmodium berghei/fisiología , Animales , Antígenos de Diferenciación de Linfocitos B/fisiología , Antígenos de Histocompatibilidad Clase II/fisiología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Hígado/inmunología , Hígado/parasitología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Malaria Cerebral/etiología , Malaria Cerebral/metabolismo , Malaria Cerebral/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Cell Microbiol ; 23(1): e13271, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32979009

RESUMEN

The protozoan parasite Plasmodium, causative agent of malaria, invades hepatocytes by invaginating the host cell plasma membrane and forming a parasitophorous vacuole membrane (PVM). Surrounded by this PVM, the parasite undergoes extensive replication. Parasites inside a PVM provoke the Plasmodium-associated autophagy-related (PAAR) response. This is characterised by a long-lasting association of the autophagy marker protein LC3 with the PVM, which is not preceded by phosphatidylinositol 3-phosphate (PI3P)-labelling. Prior to productive invasion, sporozoites transmigrate several cells and here we describe that a proportion of traversing sporozoites become trapped in a transient traversal vacuole, provoking a host cell response that clearly differs from the PAAR response. These trapped sporozoites provoke PI3P-labelling of the surrounding vacuolar membrane immediately after cell entry, followed by transient LC3-labelling and elimination of the parasite by lysosomal acidification. Our data suggest that this PI3P response is not only restricted to sporozoites trapped during transmigration but also affects invaded parasites residing in a compromised vacuole. Thus, host cells can employ a pathway distinct from the previously described PAAR response to efficiently recognise and eliminate Plasmodium parasites.


Asunto(s)
Autofagia , Hepatocitos/parasitología , Fosfatos de Fosfatidilinositol/metabolismo , Plasmodium berghei/metabolismo , Plasmodium berghei/parasitología , Esporozoítos/metabolismo , Vacuolas/parasitología , Animales , Línea Celular , Femenino , Células HeLa , Interacciones Huésped-Parásitos , Humanos , Malaria/parasitología , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Organismos Modificados Genéticamente
8.
Malar J ; 21(1): 264, 2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-36100902

RESUMEN

BACKGROUND: Sporozoites isolated from the salivary glands of Plasmodium-infected mosquitoes are a prerequisite for several basic and pre-clinical applications. Although salivary glands are pooled to maximize sporozoite recovery, insufficient yields pose logistical and analytical hurdles; thus, predicting yields prior to isolation would be valuable. Preceding oocyst densities in the midgut is an obvious candidate. However, it is unclear whether current understanding of its relationship with sporozoite densities can be used to maximize yields, or whether it can capture the potential density-dependence in rates of sporozoite invasion of the salivary glands. METHODS: This study presents a retrospective analysis of Anopheles stephensi mosquitoes infected with two strains of the rodent-specific Plasmodium berghei. Mean oocyst densities were estimated in the midguts earlier in the infection (11-15 days post-blood meal), with sporozoites pooled from the salivary glands later in the infection (17-29 days). Generalized linear mixed effects models were used to determine if (1) mean oocyst densities can predict sporozoite yields from pooled salivary glands, (2) whether these densities can capture differences in rates of sporozoite invasion of salivary glands, and (3), if the interaction between oocyst densities and time could be leveraged to boost overall yields. RESULTS: The non-linear effect of mean oocyst densities confirmed the role of density-dependent constraints in limiting yields beyond certain oocyst densities. Irrespective of oocyst densities however, the continued invasion of salivary glands by the sporozoites boosted recoveries over time (17-29 days post-blood meal) for either parasite strain. CONCLUSIONS: Sporozoite invasion of the salivary glands over time can be leveraged to maximize yields for P. berghei. In general, however, invasion of the salivary glands over time is a critical fitness determinant for all Plasmodium species (extrinsic incubation period, EIP). Thus, delaying sporozoite collection could, in principle, substantially reduce dissection effort for any parasite within the genus, with the results also alluding to the potential for changes in sporozoites densities over time to modify infectivity for the next host.


Asunto(s)
Anopheles , Esporozoítos , Animales , Anopheles/parasitología , Plasmodium berghei , Estudios Retrospectivos , Glándulas Salivales/parasitología
9.
Parasite Immunol ; 42(9): e12723, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32306409

RESUMEN

AIMS: Co-inhibitory receptors play a major role in controlling the Th1 response during blood-stage malaria. Whilst PD-1 is viewed as the dominant co-inhibitory receptor restricting T cell responses, the roles of other such receptors in coordinating Th1 cell activity during malaria are poorly understood. METHODS AND RESULTS: Here, we show that the co-inhibitory receptor Tim-3 is expressed on splenic antigen-specific T-bet+ (Th1) OT-II cells transiently during the early stage of infection with transgenic Plasmodium yoelii NL parasites expressing ovalbumin (P yoelii NL-OVA). We reveal that co-blockade of Tim-3 and PD-L1 during the acute phase of P yoelii NL infection did not improve the Th1 cell response but instead led to a specific reduction in the numbers of splenic Th1 OT-II cells. Combined blockade of Tim-3 and PD-L1 did elevate anti-parasite IgG antibody responses. Nevertheless, co-blockade of Tim-3 and PD-L1 did not affect IFN-γ production by OT-II cells and did not influence parasite control during P yoelii NL-OVA infection. CONCLUSION: Thus, our results show that Tim-3 plays an unexpected combinatorial role with PD-1 in promoting and/ or sustaining a Th1 cell response during the early phase of blood-stage P. yoelii NL infection but combined blockade does not dramatically influence anti-parasite immunity.


Asunto(s)
Receptor 2 Celular del Virus de la Hepatitis A/inmunología , Malaria/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Células TH1/inmunología , Animales , Antígeno B7-H1 , Línea Celular , Epítopos/inmunología , Malaria/parasitología , Masculino , Ratones Endogámicos C57BL , Bazo/inmunología
11.
Malar J ; 18(1): 330, 2019 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-31551073

RESUMEN

BACKGROUND: The complex life cycle of malaria parasites requires well-orchestrated stage specific gene expression. In the vertebrate host the parasites grow and multiply by schizogony in two different environments: within erythrocytes and within hepatocytes. Whereas erythrocytic parasites are well-studied in this respect, relatively little is known about the exo-erythrocytic stages. METHODS: In an attempt to fill this gap, genome wide RNA-seq analyses of various exo-erythrocytic stages of Plasmodium berghei including sporozoites, samples from a time-course of liver stage development and detached cells were performed. These latter contain infectious merozoites and represent the final step in exo-erythrocytic development. RESULTS: The analysis represents the complete transcriptome of the entire life cycle of P. berghei parasites with temporal detailed analysis of the liver stage allowing comparison of gene expression across the progression of the life cycle. These RNA-seq data from different developmental stages were used to cluster genes with similar expression profiles, in order to infer their functions. A comparison with published data from other parasite stages confirmed stage-specific gene expression and revealed numerous genes that are expressed differentially in blood and exo-erythrocytic stages. One of the most exo-erythrocytic stage-specific genes was PBANKA_1003900, which has previously been annotated as a "gametocyte specific protein". The promoter of this gene drove high GFP expression in exo-erythrocytic stages, confirming its expression profile seen by RNA-seq. CONCLUSIONS: The comparative analysis of the genome wide mRNA expression profiles of erythrocytic and different exo-erythrocytic stages could be used to improve the understanding of gene regulation in Plasmodium parasites and can be used to model exo-erythrocytic stage metabolic networks toward the identification of differences in metabolic processes during schizogony in erythrocytes and hepatocytes.


Asunto(s)
Perfilación de la Expresión Génica , Hepatocitos/parasitología , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/genética , Proteínas Protozoarias/genética , Eritrocitos/parasitología , Regulación de la Expresión Génica , Genoma de Protozoos , Humanos , Estadios del Ciclo de Vida , Hígado/parasitología , Malaria/parasitología , Merozoítos/genética , Merozoítos/crecimiento & desarrollo , Regiones Promotoras Genéticas , RNA-Seq , Esporozoítos/genética , Esporozoítos/crecimiento & desarrollo
12.
Malar J ; 18(1): 155, 2019 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-31046772

RESUMEN

BACKGROUND: The protective efficacy of the most promising malaria whole-parasite based vaccine candidates critically depends on the parasite's potential to migrate in the human host. Key components of the parasite motility machinery (e.g. adhesive proteins, actin/myosin-based motor, geometrical properties) have been identified, however the regulation of this machinery is an unknown process. METHODS: In vitro microscopic live imaging of parasites in different formulations was performed and analysed, with the quantitative analysis software SMOOTIn vitro, their motility; their adherence capacity, movement pattern and velocity during forward locomotion. RESULTS: SMOOTIn vitro enabled the detailed analysis of the regulation of the motility machinery of Plasmodium berghei in response to specific (macro)molecules in the formulation. Albumin acted as an essential supplement to induce parasite attachment and movement. Glucose, salts and other whole serum components further increased the attachment rate and regulated the velocity of the movement. CONCLUSIONS: Based on the findings can be concluded that a complex interplay of albumin, glucose and certain salts and amino acids regulates parasite motility. Insights in parasite motility regulation by supplements in solution potentially provide a way to optimize the whole-parasite malaria vaccine formulation.


Asunto(s)
Medios de Cultivo/química , Locomoción/efectos de los fármacos , Plasmodium berghei/efectos de los fármacos , Esporozoítos/fisiología , Albúminas/farmacología , Animales , Culicidae/parasitología , Medios de Cultivo/farmacología , Femenino , Glucosa/farmacología , Microscopía Intravital , Malaria/parasitología , Ratones , Plasmodium berghei/fisiología , Programas Informáticos
13.
Proc Natl Acad Sci U S A ; 113(26): 7183-8, 2016 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-27303037

RESUMEN

Transmission of the malaria parasite from the mammalian host to the mosquito vector requires the formation of adequately adapted parasite forms and stage-specific organelles. Here we show that formation of the crystalloid-a unique and short-lived organelle of the Plasmodium ookinete and oocyst stage required for sporogony-is dependent on the precisely timed expression of the S-acyl-transferase DHHC10. DHHC10, translationally repressed in female Plasmodium berghei gametocytes, is activated translationally during ookinete formation, where the protein is essential for the formation of the crystalloid, the correct targeting of crystalloid-resident protein LAP2, and malaria parasite transmission.


Asunto(s)
Aciltransferasas/fisiología , Plasmodium berghei/patogenicidad , Proteínas Protozoarias/fisiología , Animales , Femenino , Malaria/transmisión , Ratones Endogámicos BALB C , Oocistos/fisiología , Orgánulos/fisiología , Plasmodium berghei/enzimología , Plasmodium berghei/fisiología
14.
PLoS Pathog ; 12(11): e1005917, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27851824

RESUMEN

Many variant proteins encoded by Plasmodium-specific multigene families are exported into red blood cells (RBC). P. falciparum-specific variant proteins encoded by the var, stevor and rifin multigene families are exported onto the surface of infected red blood cells (iRBC) and mediate interactions between iRBC and host cells resulting in tissue sequestration and rosetting. However, the precise function of most other Plasmodium multigene families encoding exported proteins is unknown. To understand the role of RBC-exported proteins of rodent malaria parasites (RMP) we analysed the expression and cellular location by fluorescent-tagging of members of the pir, fam-a and fam-b multigene families. Furthermore, we performed phylogenetic analyses of the fam-a and fam-b multigene families, which indicate that both families have a history of functional differentiation unique to RMP. We demonstrate for all three families that expression of family members in iRBC is not mutually exclusive. Most tagged proteins were transported into the iRBC cytoplasm but not onto the iRBC plasma membrane, indicating that they are unlikely to play a direct role in iRBC-host cell interactions. Unexpectedly, most family members are also expressed during the liver stage, where they are transported into the parasitophorous vacuole. This suggests that these protein families promote parasite development in both the liver and blood, either by supporting parasite development within hepatocytes and erythrocytes and/or by manipulating the host immune response. Indeed, in the case of Fam-A, which have a steroidogenic acute regulatory-related lipid transfer (START) domain, we found that several family members can transfer phosphatidylcholine in vitro. These observations indicate that these proteins may transport (host) phosphatidylcholine for membrane synthesis. This is the first demonstration of a biological function of any exported variant protein family of rodent malaria parasites.


Asunto(s)
Hepatocitos/virología , Malaria Falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Modelos Animales de Enfermedad , Eritrocitos/parasitología , Técnica del Anticuerpo Fluorescente , Humanos , Hígado , Malaria Falciparum/virología , Ratones , Familia de Multigenes , Organismos Modificados Genéticamente , Filogenia , Plasmodium falciparum , Transporte de Proteínas , Vacuolas/virología
16.
Malar J ; 17(1): 288, 2018 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-30092798

RESUMEN

BACKGROUND: Rodent malaria parasites where the gene encoding circumsporozoite protein (CSP) has been replaced with csp genes from the human malaria parasites, Plasmodium falciparum or Plasmodium vivax, are used as pre-clinical tools to evaluate CSP vaccines in vivo. These chimeric rodent parasites produce sporozoites in Anopheles stephensi mosquitoes that are capable of infecting rodent and human hepatocytes. The availability of chimeric P. falciparum parasites where the pfcsp gene has been replaced by the pvcsp would open up possibilities to test P. vivax CSP vaccines in small scale clinical trials using controlled human malaria infection studies. METHODS: Using CRISPR/Cas9 gene editing two chimeric P. falciparum parasites, were generated, where the pfcsp gene has been replaced by either one of the two major pvcsp alleles, VK210 or VK247. In addition, a P. falciparum parasite line that lacks CSP expression was also generated. These parasite lines have been analysed for sporozoite production in An. stephensi mosquitoes. RESULTS: The two chimeric Pf-PvCSP lines exhibit normal asexual and sexual blood stage development in vitro and produce sporozoite-containing oocysts in An. stephensi mosquitoes. Expression of the corresponding PvCSP was confirmed in oocyst-derived Pf-PvCSP sporozoites. However, most oocysts degenerate before sporozoite formation and sporozoites were not found in either the mosquito haemocoel or salivary glands. Unlike the chimeric Pf-PvCSP parasites, oocysts of P. falciparum parasites lacking CSP expression do not produce sporozoites. CONCLUSIONS: Chimeric P. falciparum parasites expressing P. vivax circumsporozoite protein fail to produce salivary gland sporozoites. Combined, these studies show that while PvCSP can partially complement the function of PfCSP, species-specific features of CSP govern full sporozoite maturation and development in the two human malaria parasites.


Asunto(s)
Anopheles/parasitología , Quimera/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Esporozoítos/fisiología , Animales , Expresión Génica , Plasmodium vivax/genética , Proteínas Protozoarias/metabolismo , Glándulas Salivales/parasitología
17.
Malar J ; 17(1): 102, 2018 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-29506544

RESUMEN

BACKGROUND: Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a complication of malaria with a lethality rate of up to 80% despite anti-malarial treatment. It is characterized by a vast infiltration of leukocytes, microhaemorrhages and vasogenic oedema in the lungs. Previously, a mouse model for MA-ARDS was developed by infection of C57BL/6 mice with the Edinburgh line NK65-E of Plasmodium berghei. RESULTS: Here, both host and parasite factors were demonstrated to play crucial roles in the development and severity of lung pathology. In particular, the genetic constitution of the host was an important determinant in the development of MA-ARDS. Both male and female C57BL/6, but not BALB/c, mice developed MA-ARDS when infected with P. berghei NK65-E. However, the New York line of P. berghei NK65 (NK65-NY) did not induce demonstrable MA-ARDS, despite its accumulation in the lungs and fat tissue to a similar or even higher extent as P. berghei NK65-E. These two commonly used lines of P. berghei differ in their red blood cell preference. P. berghei NK65-NY showed a stronger predilection for reticulocytes than P. berghei NK65-E and this appeared to be associated with a lower pathogenicity in the lungs. The pulmonary pathology in the C57BL/6/P. berghei NK65-E model was more pronounced than in the model with infection of DBA/2 mice with P. berghei strain ANKA. The transient lung pathology in DBA/2 mice infected with P. berghei ANKA coincided with the infection phase in which parasites mainly infected normocytes. This phase was followed by a less pathogenic phase in which P. berghei ANKA mainly infected reticulocytes. CONCLUSIONS: The propensity of mice to develop MA-ARDS during P. berghei infection depends on both host and parasite factors and appears to correlate with RBC preference. These data provide insights in induction of MA-ARDS and may guide the choice of different mouse-parasite combinations to study lung pathology.


Asunto(s)
Modelos Animales de Enfermedad , Malaria/complicaciones , Plasmodium berghei/patogenicidad , Síndrome de Dificultad Respiratoria/patología , Animales , Femenino , Interacciones Huésped-Parásitos , Pulmón/patología , Malaria/parasitología , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
18.
Mol Microbiol ; 101(1): 78-91, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26991313

RESUMEN

Multidrug resistance (MDR) proteins belong to the B subfamily of the ATP Binding Cassette (ABC) transporters, which export a wide range of compounds including pharmaceuticals. In this study, we used reverse genetics to study the role of all seven Plasmodium MDR proteins during the life cycle of malaria parasites. Four P. berghei genes (encoding MDR1, 4, 6 and 7) were refractory to deletion, indicating a vital role during blood stage multiplication and validating them as potential targets for antimalarial drugs. Mutants lacking expression of MDR2, MDR3 and MDR5 were generated in both P. berghei and P. falciparum, indicating a dispensable role for blood stage development. Whereas P. berghei mutants lacking MDR3 and MDR5 had a reduced blood stage multiplication in vivo, blood stage growth of P. falciparum mutants in vitro was not significantly different. Oocyst maturation and sporozoite formation in Plasmodium mutants lacking MDR2 or MDR5 was reduced. Sporozoites of these P. berghei mutants were capable of infecting mice and life cycle completion, indicating the absence of vital roles during liver stage development. Our results demonstrate vital and dispensable roles of MDR proteins during blood stages and an important function in sporogony for MDR2 and MDR5 in both Plasmodium species.


Asunto(s)
Culicidae/parasitología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/metabolismo , Animales , Antimaláricos/farmacología , Resistencia a Múltiples Medicamentos , Femenino , Estadios del Ciclo de Vida , Malaria/parasitología , Malaria Falciparum/parasitología , Masculino , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Oocitos/metabolismo , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo , Plasmodium falciparum/genética , Plasmodium falciparum/crecimiento & desarrollo , Esporozoítos/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-28348156

RESUMEN

The sporogonic stage of the life cycle of Plasmodium spp., the causative agents of malaria, occurs inside the parasite's mosquito vector, where a process of fertilization, meiosis, and mitotic divisions culminates in the generation of large numbers of mammalian-infective sporozoites. Efforts to cultivate Plasmodium mosquito stages in vitro have proved challenging and yielded only moderate success. Here, we describe a methodology that simplifies the in vitro screening of much-needed transmission-blocking (TB) compounds employing a bioluminescence-based method to monitor the in vitro development of sporogonic stages of the rodent malaria parasite Plasmodium berghei Our proof-of-principle assessment of the in vitro TB activity of several commonly used antimalarial compounds identified cycloheximide, thiostrepton, and atovaquone as the most active compounds against the parasite's sporogonic stages. The TB activity of these compounds was further confirmed by in vivo studies that validated our newly developed in vitro approach to TB compound screening.


Asunto(s)
Antimaláricos/farmacología , Malaria/transmisión , Plasmodium berghei/efectos de los fármacos , Animales , Anopheles/efectos de los fármacos , Antimaláricos/uso terapéutico , Drosophila , Proteínas de Drosophila/metabolismo , Insectos Vectores/efectos de los fármacos , Malaria/tratamiento farmacológico , Esporozoítos/efectos de los fármacos
20.
Cell Microbiol ; 18(3): 369-83, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26332724

RESUMEN

Multidrug resistance-associated proteins (MRPs) belong to the C-family of ATP-binding cassette (ABC) transport proteins and are known to transport a variety of physiologically important compounds and to be involved in the extrusion of pharmaceuticals. Rodent malaria parasites encode a single ABC transporter subfamily C protein, whereas human parasites encode two: MRP1 and MRP2. Although associated with drug resistance, their biological function and substrates remain unknown. To elucidate the role of MRP throughout the parasite life cycle, Plasmodium berghei and Plasmodium falciparum mutants lacking MRP expression were generated. P. berghei mutants lacking expression of the single MRP as well as P. falciparum mutants lacking MRP1, MRP2 or both proteins have similar blood stage growth kinetics and drug-sensitivity profiles as wild type parasites. We show that MRP1-deficient parasites readily invade primary human hepatocytes and develop into mature liver stages. In contrast, both P. falciparum MRP2-deficient parasites and P. berghei mutants lacking MRP protein expression abort in mid to late liver stage development, failing to produce mature liver stages. The combined P. berghei and P. falciparum data are the first demonstration of a critical role of an ABC transporter during Plasmodium liver stage development.


Asunto(s)
Hígado/parasitología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Plasmodium berghei/patogenicidad , Plasmodium falciparum/patogenicidad , Esporozoítos/fisiología , Animales , Animales Modificados Genéticamente , Antimaláricos/farmacología , Sangre/parasitología , Femenino , Hepatocitos/parasitología , Interacciones Huésped-Parásitos , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Mutación , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA