Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
PLoS Pathog ; 12(1): e1005370, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26735921

RESUMEN

Plasmodium salivary sporozoites are the infectious form of the malaria parasite and are dormant inside salivary glands of Anopheles mosquitoes. During dormancy, protein translation is inhibited by the kinase UIS1 that phosphorylates serine 59 in the eukaryotic initiation factor 2α (eIF2α). De-phosphorylation of eIF2α-P is required for the transformation of sporozoites into the liver stage. In mammalian cells, the de-phosphorylation of eIF2α-P is mediated by the protein phosphatase 1 (PP1). Using a series of genetically knockout parasites we showed that in malaria sporozoites, contrary to mammalian cells, the eIF2α-P phosphatase is a member of the PP2C/PPM phosphatase family termed UIS2. We found that eIF2α was highly phosphorylated in uis2 conditional knockout sporozoites. These mutant sporozoites maintained the crescent shape after delivery into mammalian host and lost their infectivity. Both uis1 and uis2 were highly transcribed in the salivary gland sporozoites but uis2 expression was inhibited by the Pumilio protein Puf2. The repression of uis2 expression was alleviated when sporozoites developed into liver stage. While most eukaryotic phosphatases interact transiently with their substrates, UIS2 stably bound to phosphorylated eIF2α, raising the possibility that high-throughput searches may identify chemicals that disrupt this interaction and prevent malaria infection.


Asunto(s)
Interacciones Huésped-Parásitos/fisiología , Malaria/parasitología , Monoéster Fosfórico Hidrolasas/metabolismo , Plasmodium berghei/enzimología , Plasmodium berghei/crecimiento & desarrollo , Esporozoítos/enzimología , Esporozoítos/crecimiento & desarrollo , Animales , Línea Celular , Factor 2 Eucariótico de Iniciación/metabolismo , Técnicas de Inactivación de Genes , Humanos , Immunoblotting , Inmunoprecipitación , Estadios del Ciclo de Vida , Ratones , Fosforilación
2.
Malar J ; 14: 324, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26289816

RESUMEN

BACKGROUND: Emerging resistance of the malaria parasite Plasmodium to current therapies underscores the critical importance of exploring novel strategies for disease eradication. Plasmodium species are obligate intracellular protozoan parasites. They rely on an unusual form of substrate-dependent motility for their migration on and across host-cell membranes and for host cell invasion. This peculiar motility mechanism is driven by the 'glideosome', an actin-myosin associated, macromolecular complex anchored to the inner membrane complex of the parasite. Myosin A, actin, aldolase, and thrombospondin-related anonymous protein (TRAP) constitute the molecular core of the glideosome in the sporozoite, the mosquito stage that brings the infection into mammals. METHODS: Virtual library screening of a large compound library against the PfAldolase-TRAP complex was used to identify candidate compounds that stabilize and prevent the disassembly of the glideosome. The mechanism of these compounds was confirmed by biochemical, biophysical and parasitological methods. RESULTS: A novel inhibitory effect on the parasite was achieved by stabilizing a protein-protein interaction within the glideosome components. Compound 24 disrupts the gliding and invasive capabilities of Plasmodium parasites in in vitro parasite assays. A high-resolution, ternary X-ray crystal structure of PfAldolase-TRAP in complex with compound 24 confirms the mode of interaction and serves as a platform for future ligand optimization. CONCLUSION: This proof-of-concept study presents a novel approach to anti-malarial drug discovery and design. By strengthening a protein-protein interaction within the parasite, an avenue towards inhibiting a previously "undruggable" target is revealed and the motility motor responsible for successful invasion of host cells is rendered inactive. This study provides new insights into the malaria parasite cell invasion machinery and convincingly demonstrates that liver cell invasion is dramatically reduced by 95 % in the presence of the small molecule stabilizer compound 24.


Asunto(s)
Fructosa-Bifosfato Aldolasa/metabolismo , Proteínas de la Membrana/metabolismo , Complejos Multiproteicos/química , Proteínas Protozoarias/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Fructosa-Bifosfato Aldolasa/química , Hepatocitos/efectos de los fármacos , Humanos , Proteínas de la Membrana/química , Simulación del Acoplamiento Molecular , Complejos Multiproteicos/efectos de los fármacos , Plasmodium falciparum/química , Estabilidad Proteica/efectos de los fármacos , Proteínas Protozoarias/química , Conejos , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/toxicidad , Resonancia por Plasmón de Superficie
3.
Proc Natl Acad Sci U S A ; 109(10): 3956-61, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22355110

RESUMEN

In response to environmental stresses, the mammalian serine threonine kinases PERK, GCN2, HRI, and PKR phosphorylate the regulatory serine 51 of the eukaryotic translation initiation factor 2α (eIF2α) to inhibit global protein synthesis. Plasmodium, the protozoan that causes malaria, expresses three eIF2α kinases: IK1, IK2, and PK4. Like GCN2, IK1 regulates stress response to amino acid starvation. IK2 inhibits development of malaria sporozoites present in the mosquito salivary glands. Here we show that the phosphorylation by PK4 of the regulatory serine 59 of Plasmodium eIF2α is essential for the completion of the parasite's erythrocytic cycle that causes disease in humans. PK4 activity leads to the arrest of global protein synthesis in schizonts, where ontogeny of daughter merozoites takes place, and in gametocytes that infect Anopheles mosquitoes. The implication of these findings is that drugs that reduce PK4 activity should alleviate disease and inhibit malaria transmission.


Asunto(s)
Plasmodium falciparum/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , eIF-2 Quinasa/metabolismo , Animales , Anopheles , Codón , ADN/genética , Proteínas Fúngicas/química , Células Hep G2 , Humanos , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Mutación , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Serina/química
4.
Infect Immun ; 82(2): 793-807, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24478093

RESUMEN

Plasmodium vivax is the most widespread and the second most prevalent malaria-causing species in the world. Current measures used to control the transmission of this disease would benefit from the development of an efficacious vaccine. In the case of the deadly parasite P. falciparum, the recombinant RTS,S vaccine containing the circumsporozoite antigen (CSP) consistently protects 30 to 50% of human volunteers against infection and is undergoing phase III clinical trials in Africa with similar efficacy. These findings encouraged us to develop a P. vivax vaccine containing the three circulating allelic forms of P. vivax CSP. Toward this goal, we generated three recombinant bacterial proteins representing the CSP alleles, as well as a hybrid polypeptide called PvCSP-All-CSP-epitopes. This hybrid contains the conserved N and C termini of P. vivax CSP and the three variant repeat domains in tandem. We also generated simian and human recombinant replication-defective adenovirus vectors expressing PvCSP-All-CSP-epitopes. Mice immunized with the mixture of recombinant proteins in a formulation containing the adjuvant poly(I·C) developed high and long-lasting serum IgG titers comparable to those elicited by proteins emulsified in complete Freund's adjuvant. Antibody titers were similar in mice immunized with homologous (protein-protein) and heterologous (adenovirus-protein) vaccine regimens. The antibodies recognized the three allelic forms of CSP, reacted to the repeated and nonrepeated regions of CSP, and recognized sporozoites expressing the alleles VK210 and VK247. The vaccine formulations described in this work should be useful for the further development of an anti-P. vivax vaccine.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Vacunación/métodos , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antiprotozoarios/sangre , Femenino , Inmunoglobulina G/sangre , Vacunas contra la Malaria/administración & dosificación , Vacunas contra la Malaria/genética , Malaria Vivax/inmunología , Ratones , Ratones Endogámicos C57BL , Plasmodium vivax/genética , Poli I-C/administración & dosificación , Proteínas Protozoarias/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
5.
Eukaryot Cell ; 12(2): 161-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23243065

RESUMEN

The life cycles of apicomplexan parasites such as Plasmodium spp. and Toxoplasma gondii are complex, consisting of proliferative and latent stages within multiple hosts. Dramatic transformations take place during the cycles, and they demand precise control of gene expression at all levels, including translation. This review focuses on the mechanisms that regulate translational control in Plasmodium and Toxoplasma, with a particular emphasis on the phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eIF2α). Phosphorylation of eIF2α (eIF2α∼P) is a conserved mechanism that eukaryotic cells use to repress global protein synthesis while enhancing gene-specific translation of a subset of mRNAs. Elevated levels of eIF2α∼P have been observed during latent stages in both Toxoplasma and Plasmodium, indicating that translational control plays a role in maintaining dormancy. Parasite-specific eIF2α kinases and phosphatases are also required for proper developmental transitions and adaptation to cellular stresses encountered during the life cycle. Identification of small-molecule inhibitors of apicomplexan eIF2α kinases may selectively interfere with parasite translational control and lead to the development of new therapies to treat malaria and toxoplasmosis.


Asunto(s)
Plasmodium/genética , Biosíntesis de Proteínas , Toxoplasma/genética , Animales , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Interacciones Huésped-Parásitos , Humanos , Malaria/parasitología , Fosforilación , Plasmodium/metabolismo , Plasmodium/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Protozoarias/metabolismo , Esporozoítos/fisiología , Toxoplasma/metabolismo , Toxoplasma/fisiología , Toxoplasmosis/parasitología
6.
Proc Natl Acad Sci U S A ; 107(3): 1172-7, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20080619

RESUMEN

There is yet no licensed vaccine against malaria, a serious human disease affecting mostly children, with an annual death rate of about one million. Plasmodia, the malaria-causing parasites, have two obligatory hosts: mammals or birds, in which they multiply asexually, and mosquitoes with sexual multiplication. The most common and serious type of malaria is caused by Plasmodium falciparum. The circumsporozoite protein (CSP), a major surface antigen of sporozoites, is a protective antigen. A unique feature of P. falciparum CSP is its large central domain composed of over 30 tetrapeptide repeats of Asn-Ala-Asn-Pro (NANP). Several NANP peptide-protein conjugates were tested clinically but elicited a low level of CSP antibodies for a short duration. To provide a CSP-based candidate vaccine, we investigated recombinant CSP and NANP conjugates of various peptide lengths, with different N-terminal amino acids, bound at different ratios to various carrier proteins. Injected into mice, CSP alone and CSP or NANP conjugates induced antibodies with booster responses and were positive by the sporozoite immunofluorescent assay. The use of the mosquito stage P. falciparum ookinete surface protein, Pfs25, cross-linked onto itself as a carrier for NANP, induced in mice high levels of uniquely long-lasting antibodies to both vaccine components with secondary biological activities, that will provide immunity to liver infection by sporozoites and block transmission by mosquitoes.


Asunto(s)
Vacunas contra la Malaria/inmunología , Péptidos/química , Plasmodium falciparum/inmunología , Proteínas Protozoarias/química , Animales , Anticuerpos Antiprotozoarios/inmunología , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Femenino , Técnica del Anticuerpo Fluorescente , Cromatografía de Gases y Espectrometría de Masas , Vacunas contra la Malaria/química , Ratones , Péptidos/inmunología , Proteínas Protozoarias/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/inmunología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Linfocitos T/inmunología
7.
Nature ; 444(7121): 937-40, 2006 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-17151604

RESUMEN

Malaria infection starts when mosquitoes inject sporozoites into the skin. The parasites enter the blood stream and make their way to the liver where they develop into the exo-erythrocytic forms (EEFs). Immunization with irradiated sporozoites (IrSp) leads to robust protection against malaria infection in rodents, monkeys and humans by eliciting antibodies to circumsporozoite protein (CS) that inhibit sporozoite infectivity, and T cells that destroy the EEFs. To study the role of non-CS antigens in protection, we produced CS transgenic mice that were tolerant to CS T-cell epitopes. Here we show that in the absence of T-cell-dependent immune responses to CS, protection induced by immunization with two doses of IrSp was greatly reduced. Thus, although hundreds of other Plasmodium genes are expressed in sporozoites and EEFs, CS is a dominant protective antigen. Nevertheless, sterile immunity could be obtained by immunization of CS transgenics with three doses of IrSp.


Asunto(s)
Antígenos de Protozoos/inmunología , Epítopos Inmunodominantes/inmunología , Proteínas Protozoarias/inmunología , Esporozoítos/inmunología , Esporozoítos/efectos de la radiación , Animales , Vacunas contra la Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Esporozoítos/química , Linfocitos T Citotóxicos/inmunología
8.
J Exp Med ; 201(2): 201-9, 2005 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-15657290

RESUMEN

The yellow fever vaccine 17D (17D) is safe, and after a single immunizing dose, elicits long-lasting, perhaps lifelong protective immunity. One of the major challenges facing delivery of human vaccines in underdeveloped countries is the need for multiple injections to achieve full efficacy. To examine 17D as a vector for microbial T cell epitopes, we inserted the H-2K(d)-restricted CTL epitope of the circumsporozoite protein (CS) of Plasmodium yoelii between 17D nonstructural proteins NS2B and NS3. The recombinant virus, 17D-Py, was replication competent and stable in vitro and in vivo. A single subcutaneous injection of 10(5) PFU diminished the parasite burden in the liver by approximately 70%. The high level of protection lasted between 4 and 8 wk after immunization, but a significant effect was documented even 24 wk afterwards. Thus, the immunogenicity of a foreign T cell epitope inserted into 17D mimics some of the remarkable properties of the human vaccine. Priming with 17D-Py followed by boosting with irradiated sporozoites conferred sterile immunity to 90% of the mice. This finding indicates that the immune response of vaccine-primed individuals living in endemic areas could be sustained and magnified by the bite of infected mosquitoes.


Asunto(s)
Epítopos/inmunología , Malaria/prevención & control , Linfocitos T Citotóxicos/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Animales , Antígenos de Protozoos/inmunología , Linfocitos T CD8-positivos/inmunología , Humanos , Malaria/inmunología , Ratones , Plasmodium yoelii/inmunología , Vacuna contra la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/inmunología
9.
Mem Inst Oswaldo Cruz ; 106 Suppl 1: 167-71, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21881771

RESUMEN

Recently, we described the improved immunogenicity of new malaria vaccine candidates based on the expression of fusion proteins containing immunodominant epitopes of merozoites and Salmonella enterica serovar Typhimurium flagellin (FliC) protein as an innate immune agonist. Here, we tested whether a similar strategy, based on an immunodominant B-cell epitope from malaria sporozoites, could also generate immunogenic fusion polypeptides. A recombinant His6-tagged FliC protein containing the C-terminal repeat regions of the VK210 variant of Plasmodium vivax circumsporozoite (CS) protein was constructed. This recombinant protein was successfully expressed in Escherichia coli as soluble protein and was purified by affinity to Ni-agarose beads followed by ion exchange chromatography. A monoclonal antibody specific for the CS protein of P. vivax sporozoites (VK210) was able to recognise the purified protein. C57BL/6 mice subcutaneously immunised with the recombinant fusion protein in the absence of any conventional adjuvant developed protein-specific systemic antibody responses. However, in mice genetically deficient in expression of TLR5, this immune response was extremely low. These results extend our previous observations concerning the immunogenicity of these recombinant fusion proteins and provide evidence that the main mechanism responsible for this immune activation involves interactions with TLR5, which has not previously been demonstrated for any recombinant FliC fusion protein.


Asunto(s)
Flagelina/inmunología , Epítopos Inmunodominantes/inmunología , Vacunas contra la Malaria/inmunología , Malaria Vivax/prevención & control , Plasmodium falciparum/inmunología , Proteínas Recombinantes de Fusión/inmunología , Salmonella typhimurium/inmunología , Animales , Anticuerpos Antiprotozoarios/inmunología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito B/metabolismo , Proteínas de Escherichia coli/inmunología , Flagelina/metabolismo , Epítopos Inmunodominantes/metabolismo , Vacunas contra la Malaria/metabolismo , Malaria Vivax/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Protozoarias/inmunología , Proteínas Protozoarias/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Salmonella typhimurium/metabolismo , Receptor Toll-Like 5/inmunología
10.
Antimicrob Agents Chemother ; 54(7): 2987-93, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20457823

RESUMEN

Nitrogen-containing bisphosphonates, drugs used to treat bone resorption diseases, also have activity against a broad range of protists, including blood-stage Plasmodium spp. Here, we show that new-generation "lipophilic" bisphosphonates designed as anticancer agents that block protein prenylation also have potent activity against Plasmodium liver stages, with a high (>100) therapeutic index. Treatment of mice with the bisphosphonate BPH-715 and challenge with Plasmodium berghei sporozoites revealed complete protection (no blood-stage parasites after 28 days). There was also activity against blood-stage forms in vitro and a 4-day delay in the prepatent period in vivo. The lipophilic bisphosphonates have activity against a Plasmodium geranylgeranyl diphosphate synthase (GGPPS), as well as low nM activity against human farnesyl and geranylgeranyl diphosphate synthases. The most active inhibitor in vitro and in vivo had enzyme inhibitory activity similar to that of the other, less active compounds but was more lipophilic. Lipophilic bisphosphonates are thus promising leads for novel antimalarials that target liver-stage infection.


Asunto(s)
Antimaláricos/uso terapéutico , Difosfonatos/uso terapéutico , Hígado/fisiopatología , Plasmodium berghei/efectos de los fármacos , Animales , Difosfonatos/química , Células Hep G2 , Humanos , Ratones , Modelos Biológicos , Estructura Molecular , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/patogenicidad
11.
Hum Vaccin ; 6(1): 90-6, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19806009

RESUMEN

The RTS,S/AS01(E) malaria vaccine candidate has recently entered Phase 3 testing. Reaching this important milestone is the culmination of more than 20 years of research and development by GlaxoSmithKline and partners and collaborators. The vaccine has been developed to protect young children and infants living in Sub-Saharan Africa against clinical and severe disease caused by Plasmodium falciparum infection. Over the past 9 years, RTS,S/AS has been evaluated in multiple Phase 2 studies. The vaccine was shown to have a favorable safety profile and to be well tolerated in all age groups in which it was tested, including the intended target population of infants and young children in Sub-Saharan Africa. Data obtained so far suggest that RTS,S/AS can be co-administered with other vaccines included in the routine Expanded Program of Immunization (EPI). In Phase 2 testing, the vaccine candidate was shown to confer significant protection against P. falciparum infection and clinical disease, including severe malaria. Furthermore, a trend towards an indirect beneficial effect of the vaccine on non-malarial morbidities has been observed in several trials. In this paper, we will describe the genesis of the RTS,S/AS concept, including the rationale for selecting the circumsporozoite protein (CSP) as the target antigen. Early development history of the vaccine will be briefly described. We will present the most salient results from recent Phase 2 studies conducted in the target pediatric population, which have led to the decision to progress RTS,S/AS to Phase 3 testing. If the Phase 3 results confirm the observations made during Phase 2 testing, the RTS,S/AS vaccine, when broadly implemented and judiciously integrated with other malaria-prevention measures, would have a major public-health impact in Sub-Saharan Africa.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria Falciparum/epidemiología , Malaria Falciparum/prevención & control , África del Sur del Sahara/epidemiología , Preescolar , Ensayos Clínicos como Asunto , Humanos , Lactante , Vacunas contra la Malaria/efectos adversos , Proteínas Protozoarias/inmunología
12.
Infect Immun ; 76(3): 1200-6, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18160479

RESUMEN

The irradiated-sporozoite vaccine elicits sterile immunity against Plasmodium parasites in experimental rodent hosts and human volunteers. Based on rodent malaria models, it has been proposed that CD8+ T cells are the key protective effector mechanism required in sporozoite-induced immunity. To investigate the role of class II-restricted immunity in protective immunity, we immunized beta2-microglobulin knockout (beta2M-/-) mice with irradiated Plasmodium yoelii or P. berghei sporozoites. Sterile immunity was obtained in the CD8+-T-cell-deficient mice immunized with either P. berghei or P. yoelii sporozoites. beta2M-/- mice with the BALB/c (H-2d) genetic background as well as those with the C57BL (H-2b) genetic background were protected. Effector mechanisms included CD4+ T cells, mediated in part through the production of gamma interferon, and neutralizing antibodies that targeted the extracellular sporozoites. We conclude that in the absence of class I-restricted CD8+ T cells, sporozoite-induced protective immunity can be effectively mediated by class II-restricted immune effector mechanisms. These results support efforts to develop subunit vaccines that effectively elicit high levels of antibody and CD4+ T cells to target Plasmodium pre-erythrocytic stages.


Asunto(s)
Malaria/prevención & control , Plasmodium berghei/inmunología , Plasmodium yoelii/inmunología , Esporozoítos/inmunología , Animales , Anticuerpos Antiprotozoarios/sangre , Linfocitos T CD4-Positivos/inmunología , Interferón gamma/inmunología , Hígado/parasitología , Depleción Linfocítica , Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Plasmodium berghei/efectos de la radiación , Plasmodium yoelii/efectos de la radiación , Microglobulina beta-2/deficiencia
13.
Trends Parasitol ; 24(12): 564-9, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18929511

RESUMEN

The pre-erythrocytic (PE) phase of malaria infection, which extends from injection of sporozoites into the skin to the release of the first generation of merozoites, has traditionally been the 'black box' of the Plasmodium life cycle. However, since the advent of parasite transfection technology 13 years ago, our understanding of the PE phase in cellular and molecular terms has dramatically improved. Here, we review and comment on the major developments in the field in the past five years. Progress has been made in many diverse areas, including identifying and characterizing new proteins of interest, imaging parasites in vivo, understanding better the cell biology of hepatocyte infection and developing new vaccines against PE stages of the parasite.


Asunto(s)
Plasmodium/genética , Plasmodium/fisiología , Animales , Regulación de la Expresión Génica/fisiología , Interacciones Huésped-Parásitos/fisiología , Vacunas contra la Malaria , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
14.
Sci Rep ; 8(1): 1118, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-29348479

RESUMEN

Vaccine development against Plasmodium vivax malaria lags behind that for Plasmodium falciparum. To narrow this gap, we administered recombinant antigens based on P. vivax circumsporozoite protein (CSP) to mice. We expressed in Pichia pastoris two chimeric proteins by merging the three central repeat regions of different CSP alleles (VK210, VK247, and P. vivax-like). The first construct (yPvCSP-AllFL) contained the fused repeat regions flanked by N- and C-terminal regions. The second construct (yPvCSP-AllCT) contained the fused repeat regions and the C-terminal domain, plus RI region. Mice were vaccinated with three doses of yPvCSP in adjuvants Poly (I:C) or Montanide ISA720. We also used replication-defective adenovirus vectors expressing CSP of human serotype 5 (AdHu5) and chimpanzee serotype 68 (AdC68) for priming mice which were subsequently boosted twice with yPvCSP proteins in Poly (I:C) adjuvant. Regardless of the regime used, immunized mice generated high IgG titres specific to all CSP alleles. After challenge with P. berghei ANKA transgenic parasites expressing Pb/PvVK210 or Pb/PvVK247 sporozoites, significant time delays for parasitemia were observed in all vaccinated mice. These vaccine formulations should be clinically tried for their potential as protective universal vaccine against P. vivax malaria.


Asunto(s)
Vacunas contra la Malaria/inmunología , Malaria Vivax/inmunología , Malaria Vivax/prevención & control , Plasmodium vivax/inmunología , Proteínas Protozoarias/inmunología , Proteínas Recombinantes/inmunología , Adenoviridae/genética , Secuencia de Aminoácidos , Animales , Anticuerpos Antiprotozoarios/inmunología , Afinidad de Anticuerpos/inmunología , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/química , Inmunización , Inmunogenicidad Vacunal , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Vacunas contra la Malaria/genética , Malaria Vivax/mortalidad , Ratones , Plasmodium vivax/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética
15.
Proteins ; 66(3): 528-37, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17154157

RESUMEN

A complex molecular motor empowers substrate-dependent motility and host cell invasion in malaria parasites. The interaction between aldolase and the transmembrane adhesin thrombospondin-related anonymous protein (TRAP) transduces the motor force across the parasite surface. Here, we analyzed this interaction by using state-of-the-art flexible docking. Besides algorithms to account for induced fit in the side-chains of the Plasmodium falciparum aldolase (PfAldo) structure, we used additional in silico receptors modeled upon crystallographic structures of evolutionarily related aldolases to incorporate enzyme backbone flexibility, and to overcome structure inaccuracies due to the relatively low resolution (3.0 A) of the genuine PfAldo structure. Our results indicate that, in spite of multiple intermolecular contacts, only the six C-terminal residues of the TRAP cytoplasmic tail bind in an ordered manner to PfAldo. This portion of TRAP targets the PfAldo active site, with its n-1 Trp residue, which is essential for this interaction, buried within the PfAldo catalytic pocket. Docking of a TRAP peptide bearing a Trp to Ala mutation rendered the lower energy configurations either bound weakly outside the active site or not bound to PfAldo at all. The position of the bound TRAP peptide, and particularly the close proximity between the carbonyl of its n-2 Asp residue and the experimentally determined position of the phosphate-6 group of fructose 1,6-phosphate bound to mammalian aldolases, predicts an inhibitory effect of TRAP on catalysis. Enzymatic and TRAP-binding assays using mutant PfAldo molecules strongly support the overall structural model. These results might provide the initial framework for the identification of novel antiparasitic compounds.


Asunto(s)
Fructosa-Bifosfato Aldolasa/química , Fructosa-Bifosfato Aldolasa/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/metabolismo , Animales , Sitios de Unión , Cristalografía por Rayos X , Fructosa-Bifosfato Aldolasa/genética , Humanos , Cinética , Ligandos , Modelos Moleculares , Músculo Esquelético/enzimología , Mutación , Conejos
16.
Mol Biochem Parasitol ; 156(1): 32-40, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17714805

RESUMEN

Malaria sporozoites migrate through several cells prior to a productive invasion that involves the formation of a parasitophorous vacuole (PV) where sporozoites undergo transformation into Exo-erythorcytic forms (EEFs). The precise mechanism leading to sporozoite activation for invasion is unknown, but prior traversal of host cells is required. During cell migration sporozoites are exposed to large shifts in K(+) concentration. We report here that incubation of sporozoites to the intracellular K(+) concentration enhances 8-10 times the infectivity of Plasmodium berghei and 4-5 times the infectivity of Plasmodium yoelli sporozoites for a hepatocyte cell line, while simultaneously decreasing cell passage activity. The K(+) enhancing effect was time and concentration dependent, and was significantly decreased by K(+) channel inhibitors. Potassium-treated P. berghei sporozoites also showed enhanced numbers of EEFs in non-permissive cell lines. Treated sporozoites had reduced infectivity for mice, but infectivity was enhanced upon Kupffer cell depletion. Transcriptional analysis of K(+) treated and control sporozoites revealed a high degree of correlation in their levels of gene expression, indicating that the observed phenotypic changes are not due to radical changes in gene transcription. Only seven genes were upregulated by more than two-fold in K(+) treated sporozoites. The highest level was noted in PP2C, a phosphatase known to dephosphorylate the AKT potassium channel in plants.


Asunto(s)
Plasmodium berghei/patogenicidad , Plasmodium yoelii/patogenicidad , Potasio/farmacología , Esporozoítos/efectos de los fármacos , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Hepatocitos/parasitología , Interacciones Huésped-Parásitos , Humanos , Macrófagos del Hígado/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/crecimiento & desarrollo , Plasmodium yoelii/efectos de los fármacos , Plasmodium yoelii/crecimiento & desarrollo , Pase Seriado , Esporozoítos/crecimiento & desarrollo , Esporozoítos/fisiología
17.
Mol Biol Cell ; 14(12): 4947-57, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14595113

RESUMEN

Gliding motility and host cell invasion by apicomplexan parasites are empowered by an acto-myosin motor located underneath the parasite plasma membrane. The motor is connected to host cell receptors through trans-membrane invasins belonging to the thrombospondin-related anonymous protein (TRAP) family. A recent study indicates that aldolase bridges the cytoplasmic tail of MIC2, the homologous TRAP protein in Toxoplasma, and actin. Here, we confirm these unexpected findings in Plasmodium sporozoites and identify conserved features of the TRAP family cytoplasmic tail required to bind aldolase: a subterminal tryptophan residue and two noncontiguous stretches of negatively charged amino acids. The aldolase substrate and other compounds that bind to the active site inhibit its interaction with TRAP and with F-actin, suggesting that the function of the motor is metabolically regulated. Ultrastructural studies in salivary gland sporozoites localize aldolase to the periphery of the secretory micronemes containing TRAP. Thus, the interaction between aldolase and the TRAP tail takes place during or preceding the biogenesis of the micronemes. The release of their contents in the anterior pole of the parasite upon contact with the target cells should bring simultaneously aldolase, TRAP and perhaps F-actin to the proper subcellular location where the motor is engaged.


Asunto(s)
Fructosa-Bifosfato Aldolasa/metabolismo , Plasmodium/metabolismo , Proteínas Protozoarias/metabolismo , Esporozoítos/metabolismo , Trombospondinas/genética , Actinas/metabolismo , Secuencia de Aminoácidos , Animales , Anopheles/citología , Anopheles/metabolismo , Microscopía por Crioelectrón , Fructosa-Bifosfato Aldolasa/genética , Modelos Estructurales , Datos de Secuencia Molecular , Plasmodium/genética , Plasmodium/ultraestructura , Unión Proteica , Estructura Terciaria de Proteína/genética , Proteínas Protozoarias/genética , Glándulas Salivales/citología , Glándulas Salivales/metabolismo , Análisis de Secuencia de Proteína , Esporozoítos/genética , Esporozoítos/ultraestructura
18.
Cell Host Microbe ; 22(6): 766-776.e4, 2017 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-29241041

RESUMEN

Artemisinin and its derivatives (ARTs) are frontline antimalarial drugs. However, ART monotherapy is associated with a high frequency of recrudescent infection, resulting in treatment failure. A subset of parasites is thought to undergo ART-induced latency, but the mechanisms remain unknown. Here, we report that ART treatment results in phosphorylation of the parasite eukaryotic initiation factor-2α (eIF2α), leading to repression of general translation and latency induction. Enhanced phosphorylated eIF2α correlates with high rates of recrudescence following ART, and inhibiting eIF2α dephosphorylation renders parasites less sensitive to ART treatment. ART-induced eIF2α phosphorylation is mediated by the Plasmodium eIF2α kinase, PK4. Overexpression of a PK4 dominant-negative or pharmacological inhibition of PK4 blocks parasites from entering latency and abolishes recrudescence after ART treatment of infected mice. These results show that translational control underlies ART-induced latency and that interference with this stress response may resolve the clinical problem of recrudescent infection.


Asunto(s)
Antimaláricos/farmacología , Artemisininas/farmacología , Factor 2 Eucariótico de Iniciación/metabolismo , Plasmodium/efectos de los fármacos , Plasmodium/fisiología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Ratones , Fosforilación , Biosíntesis de Proteínas/efectos de los fármacos , Procesamiento Proteico-Postraduccional
19.
Front Immunol ; 8: 1275, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29075260

RESUMEN

Plasmodium vivax is the most common species that cause malaria outside of the African continent. The development of an efficacious vaccine would contribute greatly to control malaria. Recently, using bacterial and adenoviral recombinant proteins based on the P. vivax circumsporozoite protein (CSP), we demonstrated the possibility of eliciting strong antibody-mediated immune responses to each of the three allelic forms of P. vivax CSP (PvCSP). In the present study, recombinant proteins representing the PvCSP alleles (VK210, VK247, and P. vivax-like), as well as a hybrid polypeptide, named PvCSP-All epitopes, were generated. This hybrid containing the conserved C-terminal of the PvCSP and the three variant repeat domains in tandem were successfully produced in the yeast Pichia pastoris. After purification and biochemical characterization, they were used for the experimental immunization of C57BL/6 mice in a vaccine formulation containing the adjuvant Poly(I:C). Immunization with a recombinant protein expressing all three different allelic forms in fusion elicited high IgG antibody titers reacting with all three different allelic variants of PvCSP. The antibodies targeted both the C-terminal and repeat domains of PvCSP and recognized the native protein on the surface of P. vivax sporozoites. More importantly, mice that received the vaccine formulation were protected after challenge with chimeric Plasmodium berghei sporozoites expressing CSP repeats of P. vivax sporozoites (Pb/PvVK210). Our results suggest that it is possible to elicit protective immunity against one of the most common PvCSP alleles using soluble recombinant proteins expressed by P. pastoris. These recombinant proteins are promising candidates for clinical trials aiming to develop a multiallele vaccine against P. vivax malaria.

20.
Vaccine ; 34(38): 4501-4506, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27502569

RESUMEN

A number of studies have shown that CD8+ T cells mediate protective anti-malaria immunity in a mouse model. However, whether human CD8+ T cells play a role in protection against malaria remains unknown. We recently established human immune system (HIS) mice harboring functional human CD8+ T cells (HIS-CD8 mice) by transduction with HLA-A∗0201 and certain human cytokines using recombinant adeno-associated virus-based gene transfer technologies. These HIS-CD8 mice mount a potent, antigen-specific HLA-A∗0201-restricted human CD8+ T-cell response upon immunization with a recombinant adenovirus expressing a human malaria antigen, the Plasmodium falciparum circumsporozoite protein (PfCSP), termed AdPfCSP. In the present study, we challenged AdPfCSP-immunized HIS-CD8 mice with transgenic Plasmodium berghei sporozoites expressing full-length PfCSP and found that AdPfCSP-immunized (but not naïve) mice were protected against subsequent malaria challenge. The level of the HLA-A∗0201-restricted, PfCSP-specific human CD8+ T-cell response was closely correlated with the level of malaria protection. Furthermore, depletion of human CD8+ T cells from AdPfCSP-immunized HIS-CD8 mice almost completely abolished the anti-malaria immune response. Taken together, our data show that human CD8+ T cells mediate protective anti-malaria immunity in vivo.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra la Malaria/inmunología , Malaria Falciparum/inmunología , Malaria Falciparum/prevención & control , Animales , Dependovirus , Vectores Genéticos , Antígeno HLA-A2/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Plasmodium falciparum , Proteínas Protozoarias/inmunología , Esporozoítos
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda