Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nucleic Acids Res ; 44(13): 6087-101, 2016 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-27298255

RESUMEN

Sexual differentiation of malaria parasites into gametocytes in the vertebrate host and subsequent gamete fertilization in mosquitoes is essential for the spreading of the disease. The molecular processes orchestrating these transitions are far from fully understood. Here, we report the first transcriptome analysis of male and female Plasmodium falciparum gametocytes coupled with a comprehensive proteome analysis. In male gametocytes there is an enrichment of proteins involved in the formation of flagellated gametes; proteins involved in DNA replication, chromatin organization and axoneme formation. On the other hand, female gametocytes are enriched in proteins required for zygote formation and functions after fertilization; protein-, lipid- and energy-metabolism. Integration of transcriptome and proteome data revealed 512 highly expressed maternal transcripts without corresponding protein expression indicating large scale translational repression in P. falciparum female gametocytes for the first time. Despite a high degree of conservation between Plasmodium species, 260 of these 'repressed transcripts' have not been previously described. Moreover, for some of these genes, protein expression is only reported in oocysts and sporozoites indicating that repressed transcripts can be partitioned into short- and long-term storage. Finally, these data sets provide an essential resource for identification of vaccine/drug targets and for further mechanistic studies.


Asunto(s)
Malaria Falciparum/genética , Plasmodium falciparum/genética , Proteoma/genética , Transcriptoma/genética , Cromatina/genética , Replicación del ADN/genética , Femenino , Gametogénesis/genética , Regulación de la Expresión Génica/genética , Humanos , Malaria Falciparum/parasitología , Masculino , Redes y Vías Metabólicas/genética , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad , Biosíntesis de Proteínas , Caracteres Sexuales
2.
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
3.
Proc Natl Acad Sci U S A ; 110(19): 7862-7, 2013 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-23599283

RESUMEN

Volunteers immunized under chloroquine chemoprophylaxis with Plasmodium falciparum sporozoites (CPS) develop complete, long-lasting protection against homologous sporozoite challenge. Chloroquine affects neither sporozoites nor liver-stages, but kills only asexual forms in erythrocytes once released from the liver into the circulation. Consequently, CPS immunization exposes the host to antigens from both preerythrocytic and blood stages, and induced immunity might target either of these stages. We therefore explored the life cycle stage specificity of CPS-induced protection. Twenty-five malaria-naïve volunteers were enrolled in a clinical trial, 15 of whom received CPS immunization. Five immunized subjects and five controls received a sporozoite challenge by mosquito bites, whereas nine immunized and five control subjects received an i.v. challenge with P. falciparum-infected erythrocytes. The latter approach completely bypasses preerythrocytic stages, enabling a direct comparison of protection against either life cycle stage. CPS-immunized subjects (13 of 14) developed anticircumsporozoite antibodies, whereas only one volunteer generated minimal titers against typical blood-stage antigens. IgG from CPS-immunized volunteers did not inhibit asexual blood-stage growth in vitro. All CPS-immunized subjects (5 of 5) were protected against sporozoite challenge. In contrast, nine of nine CPS-immunized subjects developed parasitemia after blood-stage challenge, with identical prepatent periods and blood-stage multiplication rates compared with controls. Intravenously challenged CPS-immunized subjects showed earlier fever and increased plasma concentrations of inflammatory markers D-dimer, IFN-γ, and monokine induced by IFN-γ than i.v. challenged controls. The complete lack of protection against blood-stage challenge indicates that CPS-induced protection is mediated by immunity against preerythrocytic stages. However, evidence is presented for immune recognition of P. falciparum-infected erythrocytes, suggesting memory responses unable to generate functional immunity.


Asunto(s)
Cloroquina/uso terapéutico , Vacunas contra la Malaria/uso terapéutico , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Esporozoítos/inmunología , Adolescente , Adulto , Animales , Anopheles , Antígenos de Protozoos/inmunología , Antimaláricos/uso terapéutico , Eritrocitos/parasitología , Humanos , Cinética , Malaria Falciparum/tratamiento farmacológico , Resultado del Tratamiento , Adulto Joven
4.
FASEB J ; 28(5): 2158-70, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24509910

RESUMEN

The 10 Plasmodium 6-Cys proteins have critical roles throughout parasite development and are targets for antimalaria vaccination strategies. We analyzed the conserved 6-cysteine domain of this family and show that only the last 4 positionally conserved cysteine residues are diagnostic for this domain and identified 4 additional "6-Cys family-related" proteins. Two of these, sequestrin and B9, are critical to Plasmodium liver-stage development. RT-PCR and immunofluorescence assays show that B9 is translationally repressed in sporozoites and is expressed after hepatocyte invasion where it localizes to the parasite plasma membrane. Mutants lacking B9 expression in the rodent malaria parasites P. berghei and P. yoelii and the human parasite P. falciparum developmentally arrest in hepatocytes. P. berghei mutants arrest in the livers of BALB/c (100%) and C57BL6 mice (>99.9%), and in cultures of Huh7 human-hepatoma cell line. Similarly, P. falciparum mutants while fully infectious to primary human hepatocytes abort development 3 d after infection. This growth arrest is associated with a compromised parasitophorous vacuole membrane a phenotype similar to, but distinct from, mutants lacking the 6-Cys sporozoite proteins P52 and P36. Our results show that 6-Cys proteins have critical but distinct roles in establishment and maintenance of a parasitophorous vacuole and subsequent liver-stage development.


Asunto(s)
Regulación de la Expresión Génica , Hepatocitos/parasitología , Plasmodium/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Línea Celular , Biología Computacional , Cisteína/metabolismo , Femenino , Genotipo , Proteínas Fluorescentes Verdes/metabolismo , Malaria/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Mutación , Fenotipo , Plasmodium berghei/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium yoelii/metabolismo , Biosíntesis de Proteínas , Esporozoítos/crecimiento & desarrollo
5.
Eukaryot Cell ; 13(5): 550-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24297444

RESUMEN

The prodigious rate at which malaria parasites proliferate during asexual blood-stage replication, midgut sporozoite production, and intrahepatic development creates a substantial requirement for essential nutrients, including fatty acids that likely are necessary for parasite membrane formation. Plasmodium parasites obtain fatty acids either by scavenging from the vertebrate host and mosquito vector or by producing fatty acids de novo via the type two fatty acid biosynthesis pathway (FAS-II). Here, we study the FAS-II pathway in Plasmodium falciparum, the species responsible for the most lethal form of human malaria. Using antibodies, we find that the FAS-II enzyme FabI is expressed in mosquito midgut oocysts and sporozoites as well as liver-stage parasites but not during the blood stages. As expected, FabI colocalizes with the apicoplast-targeted acyl carrier protein, indicating that FabI functions in the apicoplast. We further analyze the FAS-II pathway in Plasmodium falciparum by assessing the functional consequences of deleting fabI and fabB/F. Targeted deletion or disruption of these genes in P. falciparum did not affect asexual blood-stage replication or the generation of midgut oocysts; however, subsequent sporozoite development was abolished. We conclude that the P. falciparum FAS-II pathway is essential for sporozoite development within the midgut oocyst. These findings reveal an important distinction from the rodent Plasmodium parasites P. berghei and P. yoelii, where the FAS-II pathway is known to be required for normal parasite progression through the liver stage but is not required for oocyst development in the Anopheles mosquito midgut.


Asunto(s)
Anopheles/parasitología , Ácidos Grasos/biosíntesis , Insectos Vectores/parasitología , Plasmodium falciparum/crecimiento & desarrollo , Esporozoítos/metabolismo , Animales , Tracto Gastrointestinal/parasitología , Humanos , Malaria Falciparum/parasitología , Oocistos/crecimiento & desarrollo , Oocistos/metabolismo , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Esporozoítos/crecimiento & desarrollo
6.
PLoS Pathog ; 6(4): e1000853, 2010 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-20386715

RESUMEN

The process of fertilization is critically dependent on the mutual recognition of gametes and in Plasmodium, the male gamete surface protein P48/45 is vital to this process. This protein belongs to a family of 10 structurally related proteins, the so called 6-cys family. To identify the role of additional members of this family in Plasmodium fertilisation, we performed genetic and functional analysis on the five members of the 6-cys family that are transcribed during the gametocyte stage of P. berghei. This analysis revealed that in addition to P48/45, two members (P230 and P47) also play an essential role in the process of parasite fertilization. Mating studies between parasites lacking P230, P48/45 or P47 demonstrate that P230, like P48/45, is a male fertility factor, consistent with the previous demonstration of a protein complex containing both P48/45 and P230. In contrast, disruption of P47 results in a strong reduction of female fertility, while males remain unaffected. Further analysis revealed that gametes of mutants lacking expression of p48/45 or p230 or p47 are unable to either recognise or attach to each other. Disruption of the paralog of p230, p230p, also specifically expressed in gametocytes, had no observable effect on fertilization. These results indicate that the P. berghei 6-cys family contains a number of proteins that are either male or female specific ligands that play an important role in gamete recognition and/or attachment. The implications of low levels of fertilisation that exist even in the absence of these proteins, indicating alternative pathways of fertilisation, as well as positive selection acting on these proteins, are discussed in the context of targeting these proteins as transmission blocking vaccine candidates.


Asunto(s)
Células Germinativas/metabolismo , Plasmodium berghei/fisiología , Proteínas Protozoarias/metabolismo , Animales , Secuencia de Bases , Northern Blotting , Western Blotting , Femenino , Fertilidad , Expresión Génica , Perfilación de la Expresión Génica , Masculino , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Polimorfismo Genético , Proteínas Protozoarias/genética
7.
Mol Biochem Parasitol ; 149(2): 216-22, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16824624

RESUMEN

The genome of Plasmodium falciparum contains a small gene family that expresses proteins characterized by the presence of 6-cysteine domains. Most of these proteins are expressed on the surface of the parasite and some are known to play a role in cell-cell interactions. Two members of this family, Pfs48/45 and Pfs230, form a complex localized on the surface of gametes and are recognized as important targets for transmission-blocking vaccines. In this study we report the analysis of an additional member of this family, Pfs47 the closest paralog of Pfs48/45. We demonstrate that Pfs47 is expressed only in female gametocytes and is located on the surface of female gametes following emergence from red blood cells. In contrast to the critical function of P48/45 for male fertility, Pfs47 does not appear crucial for female fertility. Parasites lacking Pfs47 through targeted gene disruption, produce normal numbers of oocysts when included in the blood meal of the mosquito vector. In addition, three monoclonal antibodies against Pfs47 were unable to inhibit oocyst development when present in a blood meal containing wild type parasites. These results show redundancy in protein function for Pfs47 and reduce the support for candidacy of Pfs47 as a transmission-blocking vaccine target.


Asunto(s)
Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Plasmodium falciparum/genética , Plasmodium falciparum/fisiología , Proteínas Protozoarias/genética , Proteínas Protozoarias/fisiología , Animales , Antígenos de Protozoos/genética , Secuencia de Bases , ADN Protozoario/genética , Femenino , Marcación de Gen , Genes Protozoarios , Células Germinativas/crecimiento & desarrollo , Masculino , Glicoproteínas de Membrana/inmunología , Oocistos/crecimiento & desarrollo , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/inmunología , Proteínas Protozoarias/inmunología
8.
Mol Biochem Parasitol ; 199(1-2): 29-33, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25827756

RESUMEN

The transmission of malaria parasites depends on the presence of sexual stages (gametocytes) in the blood, making the ratio and densities of female and male gametocytes important determinants of parasite fitness. This manuscript describes the development of reverse transcriptase quantitative PCR (RT-qPCR) assays to separately quantify mature female and male gametocytes of the human malaria parasite Plasmodium falciparum, and reveals that Pfs25 mRNA is expressed only in female gametocytes. The female (Pfs25) and male (Pfs230p) gametocyte specific RT-qPCR assays have lower detection limits of 0.3 female and 1.8 male gametocytes per microlitre of blood, respectively, making them more sensitive than microscopy. Accurate quantification of the ratio and densities of female and male gametocytes will increase understanding of P. falciparum transmission and improve the evaluation of transmission blocking interventions.


Asunto(s)
Antígenos de Protozoos/análisis , Perfilación de la Expresión Génica , Parasitología/métodos , Plasmodium falciparum/clasificación , Plasmodium falciparum/genética , Proteínas Protozoarias/análisis , Antígenos de Protozoos/genética , Proteínas Protozoarias/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Elife ; 32014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25407681

RESUMEN

A highly efficacious pre-erythrocytic stage vaccine would be an important tool for the control and elimination of malaria but is currently unavailable. High-level protection in humans can be achieved by experimental immunization with Plasmodium falciparum sporozoites attenuated by radiation or under anti-malarial drug coverage. Immunization with genetically attenuated parasites (GAP) would be an attractive alternative approach. In this study, we present data on safety and protective efficacy using sporozoites with deletions of two genes, that is the newly identified b9 and slarp, which govern independent and critical processes for successful liver-stage development. In the rodent malaria model, PbΔb9ΔslarpGAP was completely attenuated showing no breakthrough infections while efficiently inducing high-level protection. The human PfΔb9ΔslarpGAP generated without drug resistance markers were infective to human hepatocytes in vitro and to humanized mice engrafted with human hepatocytes in vivo but completely aborted development after infection. These findings support the clinical development of a PfΔb9ΔslarpSPZ vaccine.


Asunto(s)
Vacunas contra la Malaria/genética , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Esporozoítos/inmunología , Vacunas Atenuadas/genética , Animales , Humanos , Hígado/parasitología , Vacunas contra la Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plasmodium falciparum/genética , Vacunas Atenuadas/inmunología
10.
Science ; 340(6135): 984-7, 2013 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-23661646

RESUMEN

Plasmodium falciparum transmission by Anopheles gambiae mosquitoes is remarkably efficient, resulting in a very high prevalence of human malaria infection in sub-Saharan Africa. A combination of genetic mapping, linkage group selection, and functional genomics was used to identify Pfs47 as a P. falciparum gene that allows the parasite to infect A. gambiae without activating the mosquito immune system. Disruption of Pfs47 greatly reduced parasite survival in the mosquito, and this phenotype could be reverted by genetic complementation of the parasite or by disruption of the mosquito complement-like system. Pfs47 suppresses midgut nitration responses that are critical to activate the complement-like system. We provide direct experimental evidence that immune evasion mediated by Pfs47 is critical for efficient human malaria transmission by A. gambiae.


Asunto(s)
Anopheles/inmunología , Anopheles/parasitología , Malaria Falciparum/parasitología , Malaria Falciparum/transmisión , Glicoproteínas de Membrana/fisiología , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/fisiología , Animales , Técnicas de Inactivación de Genes , Humanos , Sistema Inmunológico , Glicoproteínas de Membrana/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética
11.
Vaccine ; 30(16): 2662-70, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22342550

RESUMEN

The critical first step in the clinical development of a malaria vaccine, based on live-attenuated Plasmodium falciparum sporozoites, is the guarantee of complete arrest in the liver. We report on an approach for assessing adequacy of attenuation of genetically attenuated sporozoites in vivo using the Plasmodium berghei model of malaria and P. falciparum sporozoites cultured in primary human hepatocytes. We show that two genetically attenuated sporozoite vaccine candidates, Δp52+p36 and Δfabb/f, are not adequately attenuated. Sporozoites infection of mice with both P. berghei candidates can result in blood infections. We also provide evidence that P. falciparum sporozoites of the leading vaccine candidate that is similarly attenuated through the deletion of the genes encoding the proteins P52 and P36, can develop into replicating liver stages. Therefore, we propose a minimal set of screening criteria to assess adequacy of sporozoite attenuation necessary before advancing into further clinical development and studies in humans.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria/inmunología , Plasmodium berghei/inmunología , Plasmodium falciparum/inmunología , Animales , Femenino , Eliminación de Gen , Genes Reporteros , Hepatocitos/inmunología , Hepatocitos/parasitología , Especificidad del Huésped , Humanos , Hígado/inmunología , Hígado/parasitología , Luciferasas/genética , Malaria/parasitología , Vacunas contra la Malaria/genética , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Plasmodium berghei/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Proteínas Protozoarias/inmunología , Esporozoítos/química , Esporozoítos/inmunología , Vacunas Atenuadas
12.
PLoS One ; 5(11): e15121, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-21152048

RESUMEN

Genetically-modified mutants are now indispensable Plasmodium gene-function reagents, which are also being pursued as genetically attenuated parasite vaccines. Currently, the generation of transgenic malaria-parasites requires the use of drug-resistance markers. Here we present the development of an FRT/FLP-recombinase system that enables the generation of transgenic parasites free of resistance genes. We demonstrate in the human malaria parasite, P. falciparum, the complete and efficient removal of the introduced resistance gene. We targeted two neighbouring genes, p52 and p36, using a construct that has a selectable marker cassette flanked by FRT-sequences. This permitted the subsequent removal of the selectable marker cassette by transient transfection of a plasmid that expressed a 37°C thermostable and enhanced FLP-recombinase. This method of removing heterologous DNA sequences from the genome opens up new possibilities in Plasmodium research to sequentially target multiple genes and for using genetically-modified parasites as live, attenuated malaria vaccines.


Asunto(s)
ADN Nucleotidiltransferasas/metabolismo , ADN Protozoario/metabolismo , Mutación , Plasmodium falciparum/genética , Animales , Sitios de Unión/genética , ADN Nucleotidiltransferasas/genética , ADN Protozoario/genética , Resistencia a Medicamentos/genética , Genes Protozoarios/genética , Humanos , Malaria Falciparum/parasitología , Microscopía Fluorescente , Eliminación de Secuencia , Transfección
13.
PLoS One ; 3(10): e3549, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18958160

RESUMEN

Difficulties with inducing sterile and long lasting protective immunity against malaria with subunit vaccines has renewed interest in vaccinations with attenuated Plasmodium parasites. Immunizations with sporozoites that are attenuated by radiation (RAS) can induce strong protective immunity both in humans and rodent models of malaria. Recently, in rodent parasites it has been shown that through the deletion of a single gene, sporozoites can also become attenuated in liver stage development and, importantly, immunization with these sporozoites results in immune responses identical to RAS. The promise of vaccination using these genetically attenuated sporozoites (GAS) depends on translating the results in rodent malaria models to human malaria. In this study, we perform the first essential step in this transition by disrupting, p52, in P. falciparum an ortholog of the rodent parasite gene, p36p, which we had previously shown can confer long lasting protective immunity in mice. These P. falciparum P52 deficient sporozoites demonstrate gliding motility, cell traversal and an invasion rate into primary human hepatocytes in vitro that is comparable to wild type sporozoites. However, inside the host hepatocyte development is arrested very soon after invasion. This study reveals, for the first time, that disrupting the equivalent gene in both P. falciparum and rodent malaria Plasmodium species generates parasites that become similarly arrested during liver stage development and these results pave the way for further development of GAS for human use.


Asunto(s)
Antígenos de Protozoos/genética , Marcación de Gen , Hepatocitos/parasitología , Estadios del Ciclo de Vida/genética , Hígado/parasitología , Malaria Falciparum/terapia , Plasmodium falciparum/crecimiento & desarrollo , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Culicidae/parasitología , Terapia Genética , Humanos , Malaria Falciparum/parasitología , Plasmodium berghei/genética , Plasmodium falciparum/genética , Homología de Secuencia de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda