Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Front Immunol ; 14: 1143012, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37143657

RESUMEN

Introduction: Plasmodium sporozoites (SPZ) inoculated by Anopheles mosquitoes into the skin of the mammalian host migrate to the liver before infecting hepatocytes. Previous work demonstrated that early production of IL-6 in the liver is detrimental for the parasite growth, contributing to the acquisition of a long-lasting immune protection after immunization with live attenuated parasites. Methods: Considering that IL-6 as a critical pro-inflammatory signal, we explored a novel approach whereby the parasite itself encodes for the murine IL-6 gene. We generated transgenic P. berghei parasites that express murine IL-6 during liver stage development. Results and Discussion: Though IL-6 transgenic SPZ developed into exo-erythrocytic forms in hepatocytes in vitro and in vivo, these parasites were not capable of inducing a blood stage infection in mice. Furthermore, immunization of mice with transgenic IL-6-expressing P. berghei SPZ elicited a long-lasting CD8+ T cell-mediated protective immunity against a subsequent infectious SPZ challenge. Collectively, this study demonstrates that parasite-encoded IL-6 attenuates parasite virulence with abortive liver stage of Plasmodium infection, forming the basis of a novel suicide vaccine strategy to elicit protective antimalarial immunity.


Asunto(s)
Hepatopatías , Vacunas contra la Malaria , Animales , Ratones , Linfocitos T CD8-positivos , Interleucina-6 , Mamíferos , Plasmodium berghei
2.
PLoS Pathog ; 19(3): e1011261, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36928686

RESUMEN

Invasion of host cells by apicomplexan parasites such as Toxoplasma and Plasmodium spp requires the sequential secretion of the parasite apical organelles, the micronemes and the rhoptries. The claudin-like apicomplexan microneme protein (CLAMP) is a conserved protein that plays an essential role during invasion by Toxoplasma gondii tachyzoites and in Plasmodium falciparum asexual blood stages. CLAMP is also expressed in Plasmodium sporozoites, the mosquito-transmitted forms of the malaria parasite, but its role in this stage is still unknown. CLAMP is essential for Plasmodium blood stage growth and is refractory to conventional gene deletion. To circumvent this obstacle and study the function of CLAMP in sporozoites, we used a conditional genome editing strategy based on the dimerisable Cre recombinase in the rodent malaria model parasite P. berghei. We successfully deleted clamp gene in P. berghei transmission stages and analyzed the functional consequences on sporozoite infectivity. In mosquitoes, sporozoite development and egress from oocysts was not affected in conditional mutants. However, invasion of the mosquito salivary glands was dramatically reduced upon deletion of clamp gene. In addition, CLAMP-deficient sporozoites were impaired in cell traversal and productive invasion of mammalian hepatocytes. This severe phenotype was associated with major defects in gliding motility and with reduced shedding of the sporozoite adhesin TRAP. Expansion microscopy revealed partial colocalization of CLAMP and TRAP in a subset of micronemes, and a distinct accumulation of CLAMP at the apical tip of sporozoites. Collectively, these results demonstrate that CLAMP is essential across invasive stages of the malaria parasite, and support a role of the protein upstream of host cell invasion, possibly by regulating the secretion or function of adhesins in Plasmodium sporozoites.


Asunto(s)
Culicidae , Malaria , Animales , Esporozoítos/metabolismo , Micronema , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Culicidae/parasitología , Mamíferos , Malaria/parasitología
3.
iScience ; 26(2): 106056, 2023 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-36761022

RESUMEN

Plasmodium sporozoites are transmitted to a mammalian host during blood feeding by an infected mosquito and invade hepatocytes for initial replication of the parasite into thousands of erythrocyte-invasive merozoites. Here we report that the B9 protein, a member of the 6-cysteine domain protein family, is secreted from sporozoite micronemes and is required for productive invasion of hepatocytes. The N-terminus of B9 forms a beta-propeller domain structurally related to CyRPA, a cysteine-rich protein forming an essential invasion complex in Plasmodium falciparum merozoites. The beta-propeller domain of B9 is essential for sporozoite infectivity and interacts with the 6-cysteine proteins P36 and P52 in a heterologous expression system. Our results suggest that, despite using distinct sets of parasite and host entry factors, Plasmodium sporozoites and merozoites may share common structural modules to assemble protein complexes for invasion of host cells.

4.
Open Biol ; 12(8): 220015, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35920043

RESUMEN

Protein phosphatase 1 (PP1) is a key enzyme for Plasmodium development. However, the detailed mechanisms underlying its regulation remain to be deciphered. Here, we report the functional characterization of the Plasmodium berghei leucine-rich repeat protein 1 (PbLRR1), an orthologue of SDS22, one of the most ancient and conserved PP1 interactors. Our study shows that PbLRR1 is expressed during intra-erythrocytic development of the parasite, and up to the zygote stage in mosquitoes. PbLRR1 can be found in complex with PbPP1 in both asexual and sexual stages and inhibits its phosphatase activity. Genetic analysis demonstrates that PbLRR1 depletion adversely affects the development of oocysts. PbLRR1 interactome analysis associated with phospho-proteomics studies identifies several novel putative PbLRR1/PbPP1 partners. Some of these partners have previously been characterized as essential for the parasite sexual development. Interestingly, and for the first time, Inhibitor 3 (I3), a well-known and direct interactant of Plasmodium PP1, was found to be drastically hypophosphorylated in PbLRR1-depleted parasites. These data, along with the detection of I3 with PP1 in the LRR1 interactome, strongly suggest that the phosphorylation status of PbI3 is under the control of the PP1-LRR1 complex and could contribute (in)directly to oocyst development. This study provides new insights into previously unrecognized PbPP1 fine regulation of Plasmodium oocyst development through its interaction with PbLRR1.


Asunto(s)
Proteínas Repetidas Ricas en Leucina , Plasmodium berghei , Animales , Oocistos/metabolismo , Fosforilación , Plasmodium berghei/genética , Plasmodium berghei/metabolismo , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo
5.
Front Cell Infect Microbiol ; 12: 901253, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35782147

RESUMEN

Malaria, an infection caused by apicomplexan parasites of the genus Plasmodium, continues to exact a significant toll on public health with over 200 million cases world-wide, and annual deaths in excess of 600,000. Considerable progress has been made to reduce malaria burden in endemic countries in the last two decades. However, parasite and mosquito resistance to frontline chemotherapies and insecticides, respectively, highlights the continuing need for the development of safe and effective vaccines. Here we describe the development of recombinant human antibodies to three target proteins from Plasmodium falciparum: reticulocyte binding protein homologue 5 (PfRH5), cysteine-rich protective antigen (PfCyRPA), and circumsporozoite protein (PfCSP). All three proteins are key targets in the development of vaccines for blood-stage or pre-erythrocytic stage infections. We have developed potent anti-PfRH5, PfCyRPA and PfCSP monoclonal antibodies that will prove useful tools for the standardisation of assays in preclinical research and the assessment of these antigens in clinical trials. We have generated some very potent anti-PfRH5 and anti-PfCyRPA antibodies with some clones >200 times more potent than the polyclonal anti-AMA-1 antibodies used for the evaluation of blood stage antigens. While the monoclonal and polyclonal antibodies are not directly comparable, the data provide evidence that these new antibodies are very good at blocking invasion. These antibodies will therefore provide a valuable resource and have potential as biological standards to help harmonise pre-clinical malaria research.


Asunto(s)
Anticuerpos Monoclonales , Plasmodium falciparum , Animales , Anticuerpos Antiprotozoarios , Proteínas Portadoras , Eritrocitos , Humanos
6.
Front Cell Infect Microbiol ; 12: 883759, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35694548

RESUMEN

The persistence of erythrocytes infected with Plasmodium falciparum gametocytes in the bloodstream is closely related to the modulation of their mechanical properties. New drugs that increase the stiffness of infected erythrocytes may thus represent a novel approach to block malaria parasite transmission. The phosphodiesterase inhibitor tadalafil has been shown to impair the ability of infected erythrocytes to circulate in an in vitro model for splenic retention. Here, we used a humanized mouse model to address in vivo the effect of tadalafil on the circulation kinetics of mature gametocyte-infected erythrocytes. We show that stiff immature gametocyte-infected erythrocytes are retained in the spleen of humanized mice at rates comparable to that of the in vitro model. Accordingly, tadalafil-induced stiffening of mature gametocyte-infected erythrocytes impairs their circulation in the bloodstream and triggers their retention by the spleen. These in vivo results validate that tadalafil is a novel drug lead potentially capable of blocking malaria parasite transmission by targeting GIE mechanical properties.


Asunto(s)
Malaria Falciparum , Plasmodium falciparum , Animales , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Ratones , Inhibidores de Fosfodiesterasa , Bazo , Tadalafilo/farmacología
7.
PLoS Pathog ; 18(6): e1010643, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35731833

RESUMEN

Plasmodium sporozoites that are transmitted by blood-feeding female Anopheles mosquitoes invade hepatocytes for an initial round of intracellular replication, leading to the release of merozoites that invade and multiply within red blood cells. Sporozoites and merozoites share a number of proteins that are expressed by both stages, including the Apical Membrane Antigen 1 (AMA1) and the Rhoptry Neck Proteins (RONs). Although AMA1 and RONs are essential for merozoite invasion of erythrocytes during asexual blood stage replication of the parasite, their function in sporozoites was still unclear. Here we show that AMA1 interacts with RONs in mature sporozoites. By using DiCre-mediated conditional gene deletion in P. berghei, we demonstrate that loss of AMA1, RON2 or RON4 in sporozoites impairs colonization of the mosquito salivary glands and invasion of mammalian hepatocytes, without affecting transcellular parasite migration. Three-dimensional electron microscopy data showed that sporozoites enter salivary gland cells through a ring-like structure and by forming a transient vacuole. The absence of a functional AMA1-RON complex led to an altered morphology of the entry junction, associated with epithelial cell damage. Our data establish that AMA1 and RONs facilitate host cell invasion across Plasmodium invasive stages, and suggest that sporozoites use the AMA1-RON complex to efficiently and safely enter the mosquito salivary glands to ensure successful parasite transmission. These results open up the possibility of targeting the AMA1-RON complex for transmission-blocking antimalarial strategies.


Asunto(s)
Anopheles , Plasmodium , Animales , Femenino , Anopheles/parasitología , Mamíferos , Merozoítos/metabolismo , Plasmodium/metabolismo , Plasmodium berghei/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo
8.
Mol Microbiol ; 117(3): 618-631, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34564906

RESUMEN

Apicomplexan parasites encompass diverse pathogens for humans and animals, including the causative agents of malaria and toxoplasmosis, Plasmodium spp. and Toxoplasma gondii. Genetic manipulation of these parasites has become central to explore parasite biology, unravel gene function and identify new targets for therapeutic strategies. Tremendous progress has been achieved over the past years with the advent of next generation sequencing and powerful genome editing methods. In particular, various methods for conditional gene expression have been developed in both Plasmodium and Toxoplasma to knockout or knockdown essential genes, or for inducible expression of master developmental regulators or mutant versions of proteins. Conditional gene expression can be achieved at three distinct levels. At the DNA level, inducible site-specific recombinases allow conditional genome editing. At the RNA level, regulation can be achieved during transcription, using stage-specific or regulatable promoters, or post-transcriptionally through alteration of mRNA stability or translation. At the protein level, several systems have been developed for inducible degradation or displacement of a protein of interest. In this review, we provide an overview of current systems for conditional control of gene expression in Plasmodium and Toxoplasma parasites, highlighting the advantages and limitations of each approach.


Asunto(s)
Parásitos , Plasmodium , Toxoplasma , Animales , Expresión Génica , Genes Esenciales , Parásitos/genética , Plasmodium/genética , Toxoplasma/genética
9.
Parasite Immunol ; 43(12): e12877, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34515999

RESUMEN

Sporozoite antigens are the basis of a number of malaria vaccines being tested, but the contribution of antigens expressed during subsequent liver stage development to pre-erythrocytic stage immunity is poorly understood. We previously showed that, following immunisation with radiation attenuated sporozoites (RAS), a model epitope embedded in a sporozoite surface protein elicited robust CD8+ T cell responses, whilst the same epitope in a liver stage antigen induced inferior responses. Since RAS arrest early in their development in host hepatocytes, we hypothesised that extending parasite maturation in the liver could considerably improve the epitope-specific CD8+ T cell response. Here, we employed a late liver stage arrested parasite model, azithromycin prophylaxis alongside live sporozoites, to increase expression of the model epitope until full liver stage maturation. Strikingly, this alternative immunisation strategy, which has been shown to elicit superior protection, failed to improve the resulting epitope-specific CD8+ T cell responses. Our findings support the notion that liver stage antigens are poorly immunogenic and provide additional caution about prioritising antigens for vaccine development based solely on immunogenicity.


Asunto(s)
Vacunas contra la Malaria , Plasmodium berghei , Animales , Antígenos de Protozoos , Linfocitos T CD8-positivos , Hígado/parasitología , Esporozoítos
10.
Methods Mol Biol ; 2369: 101-120, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34313986

RESUMEN

Genome editing in the malaria parasite Plasmodium relies on homologous recombination and requires parasite transfection in asexual blood stages. Therefore, conditional genetic approaches are needed to delete genes that are essential during blood stage replication. Among these, the dimerizable Cre (DiCre) recombinase system has emerged as a powerful approach for conditional gene knockout in Plasmodium parasites. In this system, the Cre recombinase is expressed in the form of two separate, enzymatically inactive polypeptides. Rapamycin-induced heterodimerization of the two components restores recombinase activity, leading to site-specific excision of floxed DNA sequences. Here, we describe methods to generate genetically modified DiCre-expressing Plasmodium berghei mutants by introducing Lox sites upstream and downstream of a gene of interest and to induce conditional excision of the floxed gene in different stages of the parasite life cycle. Administration of rapamycin to P. berghei-infected mice allows conditional gene deletion in the asexual erythrocytic stages. Rapamycin-induced gene excision can also be achieved in P. berghei sexual blood stages prior to transmission to mosquitoes, or during sporogony by treating P. berghei-infected mosquitoes, both methods allowing functional studies in P. berghei mosquito stages. Finally, rapamycin can be administered to in vitro cell cultures in order to induce gene excision in P. berghei liver stages. Subsequent phenotyping allows for the analysis of essential gene function across the parasite life cycle stages.


Asunto(s)
Culicidae , Plasmodium berghei , Animales , Eliminación de Gen , Integrasas/genética , Estadios del Ciclo de Vida , Ratones , Plasmodium berghei/genética , Sirolimus/farmacología
11.
EMBO Mol Med ; 13(4): e13390, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33709544

RESUMEN

Immunogenicity is considered one important criterion for progression of candidate vaccines to further clinical evaluation. We tested this assumption in an infection and vaccination model for malaria pre-erythrocytic stages. We engineered Plasmodium berghei parasites that harbour a well-characterised epitope for stimulation of CD8+ T cells, either as an antigen in the sporozoite surface-expressed circumsporozoite protein or the parasitophorous vacuole membrane associated protein upregulated in sporozoites 4 (UIS4) expressed in exo-erythrocytic forms (EEFs). We show that the antigen origin results in profound differences in immunogenicity with a sporozoite antigen eliciting robust, superior antigen-specific CD8+ T-cell responses, whilst an EEF antigen evokes poor responses. Despite their contrasting immunogenic properties, both sporozoite and EEF antigens gain access to antigen presentation pathways in hepatocytes, as recognition and targeting by vaccine-induced effector CD8+ T cells results in high levels of protection when targeting either antigen. Our study is the first demonstration that poorly immunogenic EEF antigens do not preclude their susceptibility to antigen-specific CD8+ T-cell killing, which has wide-ranging implications on antigen prioritisation for next-generation pre-erythrocytic malaria vaccines.


Asunto(s)
Vacunas contra la Malaria , Malaria , Animales , Linfocitos T CD8-positivos , Malaria/prevención & control , Esporozoítos , Vacunación
12.
Front Cell Infect Microbiol ; 11: 624945, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33747980

RESUMEN

Colonization of the mosquito host by Plasmodium parasites is achieved by sexually differentiated gametocytes. Gametocytogenesis, gamete formation and fertilization are tightly regulated processes, and translational repression is a major regulatory mechanism for stage conversion. Here, we present a characterization of a Plasmodium berghei RNA binding protein, UIS12, that contains two conserved eukaryotic RNA recognition motifs (RRM). Targeted gene deletion resulted in viable parasites that replicate normally during blood infection, but form fewer gametocytes. Upon transmission to Anopheles stephensi mosquitoes, both numbers and size of midgut-associated oocysts were reduced and their development stopped at an early time point. As a consequence, no salivary gland sporozoites were formed indicative of a complete life cycle arrest in the mosquito vector. Comparative transcript profiling in mutant and wild-type infected red blood cells revealed a decrease in transcript abundance of mRNAs coding for signature gamete-, ookinete-, and oocyst-specific proteins in uis12(-) parasites. Together, our findings indicate multiple roles for UIS12 in regulation of gene expression after blood infection in good agreement with the pleiotropic defects that terminate successful sporogony and onward transmission to a new vertebrate host.


Asunto(s)
Anopheles , Plasmodium berghei , Animales , Oocistos , Proteínas de Unión al ARN , Esporozoítos
13.
Proteomics ; 21(6): e2000305, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33452840

RESUMEN

Sporozoites of the malaria parasite Plasmodium are transmitted by mosquitoes and infect the liver for an initial and obligatory round of replication, before exponential multiplication in the blood and onset of the disease. Sporozoites and liver stages provide attractive targets for malaria vaccines and prophylactic drugs. In this context, defining the parasite proteome is important to explore the parasite biology and to identify potential targets for antimalarial strategies. Previous studies have determined the total proteome of sporozoites from the two main human malaria parasites, P. falciparum and P. vivax, as well as P. yoelii, which infects rodents. Another murine malaria parasite, P. berghei, is widely used to investigate the parasite biology. However, a deep view of the proteome of P. berghei sporozoites is still missing. To fill this gap, we took advantage of the highly sensitive timsTOF PRO mass spectrometer, combined with three alternative methods for sporozoite purification, to identify the proteome of P. berghei sporozoites using low numbers of parasites. This study provides a reference proteome for P. berghei sporozoites, identifying a core set of proteins expressed across species, and illustrates how the unprecedented sensitivity of the timsTOF PRO system enables deep proteomic analysis from limited sample amounts.


Asunto(s)
Plasmodium berghei , Esporozoítos , Animales , Espectrometría de Movilidad Iónica , Ratones , Proteoma , Proteómica
14.
Mol Microbiol ; 115(5): 870-881, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33191548

RESUMEN

Parasites of the genus Plasmodium, the etiological agent of malaria, are transmitted through the bite of anopheline mosquitoes, which deposit sporozoites into the host skin. Sporozoites migrate through the dermis, enter the bloodstream, and rapidly traffic to the liver. They cross the liver sinusoidal barrier and traverse several hepatocytes before switching to productive invasion of a final one for replication inside a parasitophorous vacuole. Cell traversal and productive invasion are functionally independent processes that require proteins secreted from specialized secretory organelles known as micronemes. In this review, we summarize the current understanding of how sporozoites traverse through cells and productively invade hepatocytes, and discuss the role of environmental sensing in switching from a migratory to an invasive state. We propose that timely controlled secretion of distinct microneme subsets could play a key role in successful migration and infection of hepatocytes. A better understanding of these essential biological features of the Plasmodium sporozoite may contribute to the development of new strategies to fight against the very first and asymptomatic stage of malaria.


Asunto(s)
Hepatocitos/parasitología , Malaria/parasitología , Plasmodium/fisiología , Esporozoítos/fisiología , Animales , Humanos , Hígado/parasitología , Plasmodium/genética , Plasmodium/crecimiento & desarrollo , Esporozoítos/genética , Esporozoítos/crecimiento & desarrollo
15.
J Immunol ; 205(11): 3071-3082, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33148715

RESUMEN

Malaria is associated with complicated immunopathogenesis. In this study, we provide evidence for an unexpected role of TLR3 in promoting the establishment of Plasmodium yoelii infection through delayed clearance of parasitemia in wild type C57BL/6jRj (B6) compared with TLR3 knockout mice. In this study, we confirmed an increased expression of Tlr3, Trif, Tbk1, and Irf7/Irf3 in the liver 42 h postinfection and the initiation of an early burst of proinflammatory response such as Ifng, NF-kB, and Tnfa in B6 mice that may promote parasite fitness. Interestingly, in the absence of TLR3, we showed the involvement of high IFN-γ and lower type I IFN response in the early clearance of parasitemia. In parallel, we observed an increase in splenic NK and NKT cells expressing TLR3 in infected B6 mice, suggesting a role for TLR sensing in the innate immune response. Finally, we find evidence that the increase in the frequency of CD19+TLR3+ B cells along with reduced levels of total IgG in B6 mice possibly suggests the initiation of TLR3-dependent pathway early during P. yoelii infection. Our results thus reveal a new mechanism in which a parasite-activated TLR3 pathway promotes blood stage infection along with quantitative and qualitative differences in Ab responses.


Asunto(s)
Malaria/inmunología , Mamíferos/inmunología , Mamíferos/parasitología , Plasmodium yoelii/inmunología , Receptor Toll-Like 3/inmunología , Animales , Linfocitos B/inmunología , Inmunidad Innata/inmunología , Inmunoglobulina G/inmunología , Inflamación/inmunología , Inflamación/parasitología , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/parasitología , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/inmunología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/parasitología , Parasitemia/inmunología , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/inmunología
16.
PLoS One ; 15(10): e0236616, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33044964

RESUMEN

Asexual blood stages of the malaria parasite are readily amenable to genetic modification via homologous recombination, allowing functional studies of parasite genes that are not essential in this part of the life cycle. However, conventional reverse genetics cannot be applied for the functional analysis of genes that are essential during asexual blood-stage replication. Various strategies have been developed for conditional mutagenesis of Plasmodium, including recombinase-based gene deletion, regulatable promoters, and mRNA or protein destabilization systems. Among these, the dimerisable Cre (DiCre) recombinase system has emerged as a powerful approach for conditional gene deletion in P. falciparum. In this system, the bacteriophage Cre is expressed in the form of two separate, enzymatically inactive polypeptides, each fused to a different rapamycin-binding protein. Rapamycin-induced heterodimerization of the two components restores recombinase activity. We have implemented the DiCre system in the rodent malaria parasite P. berghei, and show that rapamycin-induced excision of floxed DNA sequences can be achieved with very high efficiency in both mammalian and mosquito parasite stages. This tool can be used to investigate the function of essential genes not only in asexual blood stages, but also in other parts of the malaria parasite life cycle.


Asunto(s)
Eliminación de Gen , Edición Génica , Genes Protozoarios/genética , Integrasas/metabolismo , Malaria/parasitología , Mutagénesis , Plasmodium berghei/genética , Animales , Femenino , Integrasas/química , Integrasas/genética , Estadios del Ciclo de Vida , Malaria/genética , Malaria/metabolismo , Ratones , Ratones Endogámicos C57BL
17.
Sci Rep ; 10(1): 13509, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32782257

RESUMEN

Sporozoite forms of the Plasmodium parasite, the causative agent of malaria, are transmitted by mosquitoes and first infect the liver for an initial round of replication before parasite proliferation in the blood. The molecular mechanisms involved during sporozoite invasion of hepatocytes remain poorly understood. Two receptors of the Hepatitis C virus (HCV), the tetraspanin CD81 and the scavenger receptor class B type 1 (SR-B1), play an important role during the entry of Plasmodium sporozoites into hepatocytes. In contrast to HCV entry, which requires both CD81 and SR-B1 together with additional host factors, CD81 and SR-B1 operate independently during malaria liver infection. Sporozoites from human-infecting P. falciparum and P. vivax rely respectively on CD81 or SR-B1. Rodent-infecting P. berghei can use SR-B1 to infect host cells as an alternative pathway to CD81, providing a tractable model to investigate the role of SR-B1 during Plasmodium liver infection. Here we show that mouse SR-B1 is less functional as compared to human SR-B1 during P. berghei infection. We took advantage of this functional difference to investigate the structural determinants of SR-B1 required for infection. Using a structure-guided strategy and chimeric mouse/human SR-B1 constructs, we could map the functional region of human SR-B1 within apical loops, suggesting that this region of the protein may play a crucial role for interaction of sporozoite ligands with host cells and thus the very first step of Plasmodium infection.


Asunto(s)
Antígenos CD36/metabolismo , Hepatocitos/metabolismo , Hepatocitos/parasitología , Plasmodium/fisiología , Esporozoítos/fisiología , Secuencia de Aminoácidos , Animales , Antígenos CD36/química , Humanos , Ratones , Modelos Moleculares , Dominios Proteicos , Tetraspanina 28/metabolismo
18.
Sci Rep ; 10(1): 11379, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32647135

RESUMEN

Vector control programmes are a strategic priority in the fight against malaria. However, vector control interventions require rigorous monitoring. Entomological tools for characterizing malaria transmission drivers are limited and are difficult to establish in the field. To predict Anopheles drivers of malaria transmission, such as mosquito age, blood feeding and Plasmodium infection, we evaluated artificial neural networks (ANNs) coupled to matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry (MS) and analysed the impact on the proteome of laboratory-reared Anopheles stephensi mosquitoes. ANNs were sensitive to Anopheles proteome changes and specifically recognized spectral patterns associated with mosquito age (0-10 days, 11-20 days and 21-28 days), blood feeding and P. berghei infection, with best prediction accuracies of 73%, 89% and 78%, respectively. This study illustrates that MALDI-TOF MS coupled to ANNs can be used to predict entomological drivers of malaria transmission, providing potential new tools for vector control. Future studies must assess the field validity of this new approach in wild-caught adult Anopheles. A similar approach could be envisaged for the identification of blood meal source and the detection of insecticide resistance in Anopheles and to other arthropods and pathogens.


Asunto(s)
Anopheles/parasitología , Seguimiento de Parámetros Ecológicos/métodos , Malaria/transmisión , Mosquitos Vectores/parasitología , Proteómica/métodos , Animales , Anopheles/fisiología , Conducta Alimentaria , Femenino , Humanos , Malaria/diagnóstico , Malaria/parasitología , Malaria/prevención & control , Control de Mosquitos , Mosquitos Vectores/fisiología , Redes Neurales de la Computación , Plasmodium berghei/aislamiento & purificación , Plasmodium berghei/patogenicidad , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
19.
Infect Immun ; 88(10)2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32719159

RESUMEN

The circumsporozoite protein (CSP) builds up the surface coat of sporozoites and is the leading malaria pre-erythrocytic-stage vaccine candidate. CSP has been shown to induce robust CD8+ T cell responses that are capable of eliminating developing parasites in hepatocytes, resulting in protective immunity. In this study, we characterized the importance of the immunodominant CSP-derived epitope SYIPSAEKI of Plasmodium berghei in both sporozoite- and vaccine-induced protection in murine infection models. In BALB/c mice, where SYIPSAEKI is efficiently presented in the context of the major histocompatibility complex class I (MHC-I) molecule H-2-Kd, we established that epitope-specific CD8+ T cell responses contribute to parasite killing following sporozoite immunization. Yet, sterile protection was achieved in the absence of this epitope, substantiating the concept that other antigens can be sufficient for parasite-induced protective immunity. Furthermore, we demonstrated that SYIPSAEKI-specific CD8+ T cell responses elicited by viral-vectored CSP-expressing vaccines effectively targeted parasites in hepatocytes. The resulting sterile protection strictly relied on the expression of SYIPSAEKI. In C57BL/6 mice, which are unable to present the immunodominant epitope, CSP-based vaccines did not confer complete protection, despite the induction of high levels of CSP-specific antibodies. These findings underscore the significance of CSP in protection against malaria pre-erythrocytic stages and demonstrate that a significant proportion of the protection against the parasite is mediated by CD8+ T cells specific for the immunodominant CSP-derived epitope.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Vacunas contra la Malaria/inmunología , Malaria/prevención & control , Plasmodium berghei/inmunología , Proteínas Protozoarias/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Anticuerpos Antiprotozoarios/inmunología , Presentación de Antígeno , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Modelos Animales de Enfermedad , Epítopos de Linfocito T/química , Inmunización , Malaria/inmunología , Malaria/parasitología , Vacunas contra la Malaria/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Fragmentos de Péptidos , Proteínas Protozoarias/química , Especificidad de la Especie , Esporozoítos/inmunología
20.
Front Cell Infect Microbiol ; 10: 618430, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33585284

RESUMEN

Plasmodium sporozoites are transmitted to mammals by anopheline mosquitoes and first infect the liver, where they transform into replicative exoerythrocytic forms, which subsequently release thousands of merozoites that invade erythrocytes and initiate the malaria disease. In some species, sporozoites can transform into dormant hypnozoites in the liver, which cause malaria relapses upon reactivation. Transmission from the insect vector to a mammalian host is a critical step of the parasite life cycle, and requires tightly regulated gene expression. Sporozoites are formed inside oocysts in the mosquito midgut and become fully infectious after colonization of the insect salivary glands, where they remain quiescent until transmission. Parasite maturation into infectious sporozoites is associated with reprogramming of the sporozoite transcriptome and proteome, which depends on multiple layers of transcriptional and post-transcriptional regulatory mechanisms. An emerging scheme is that gene expression in Plasmodium sporozoites is controlled by alternating waves of transcription activity and translational repression, which shape the parasite RNA and protein repertoires for successful transition from the mosquito vector to the mammalian host.


Asunto(s)
Anopheles , Malaria , Plasmodium , Animales , Regulación de la Expresión Génica , Insectos Vectores , Plasmodium/genética , Proteínas Protozoarias/genética , Esporozoítos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...