Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 12(7): e0181656, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28732045

RESUMEN

Malaria is caused by five different Plasmodium spp. in humans each of which modifies the host erythrocyte to survive and replicate. The two main causes of malaria, P. falciparum and P. vivax, differ in their ability to cause severe disease, mainly due to differences in the cytoadhesion of infected erythrocytes (IE) in the microvasculature. Cytoadhesion of P. falciparum in the brain leads to a large number of deaths each year and is a consequence of exported parasite proteins, some of which modify the erythrocyte cytoskeleton while others such as PfEMP1 project onto the erythrocyte surface where they bind to endothelial cells. Here we investigate the effects of knocking out an exported Hsp70-type chaperone termed Hsp70-x that is present in P. falciparum but not P. vivax. Although the growth of Δhsp70-x parasites was unaffected, the export of PfEMP1 cytoadherence proteins was delayed and Δhsp70-x IE had reduced adhesion. The Δhsp70-x IE were also more rigid than wild-type controls indicating changes in the way the parasites modified their host erythrocyte. To investigate the cause of this, transcriptional and translational changes in exported and chaperone proteins were monitored and some changes were observed. We propose that PfHsp70-x is not essential for survival in vitro, but may be required for the efficient export and functioning of some P. falciparum exported proteins.


Asunto(s)
Proteínas HSP70 de Choque Térmico/metabolismo , Malaria Falciparum/parasitología , Chaperonas Moleculares/metabolismo , Plasmodium falciparum/metabolismo , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Virulencia/fisiología , Animales , Adhesión Celular/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/parasitología , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/parasitología , Malaria Falciparum/sangre , Malaria Falciparum/metabolismo , Parásitos/metabolismo , Parásitos/patogenicidad
2.
Sci Rep ; 6: 37502, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27874068

RESUMEN

Plasmodium parasites are responsible for the devastating disease malaria that affects hundreds of millions of people each year. Blood stage parasites establish new permeability pathways (NPPs) in infected red blood cell membranes to facilitate the uptake of nutrients and removal of parasite waste products. Pharmacological inhibition of the NPPs is expected to lead to nutrient starvation and accumulation of toxic metabolites resulting in parasite death. Here, we have screened a curated library of antimalarial compounds, the MMV Malaria Box, identifying two compounds that inhibit NPP function. Unexpectedly, metabolic profiling suggested that both compounds also inhibit dihydroorotate dehydrogense (DHODH), which is required for pyrimidine synthesis and is a validated drug target in its own right. Expression of yeast DHODH, which bypasses the need for the parasite DHODH, increased parasite resistance to these compounds. These studies identify two potential candidates for therapeutic development that simultaneously target two essential pathways in Plasmodium, NPP and DHODH.


Asunto(s)
Antimaláricos/farmacología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Inhibidores Enzimáticos/análisis , Inhibidores Enzimáticos/farmacología , Estadios del Ciclo de Vida/efectos de los fármacos , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Plasmodium falciparum/enzimología , Plasmodium falciparum/crecimiento & desarrollo , Animales , Antimaláricos/análisis , Antimaláricos/química , Proliferación Celular/efectos de los fármacos , Dihidroorotato Deshidrogenasa , Evaluación Preclínica de Medicamentos , Complejo III de Transporte de Electrones/metabolismo , Inhibidores Enzimáticos/química , Eritrocitos/efectos de los fármacos , Eritrocitos/parasitología , Genes Reporteros , Ensayos Analíticos de Alto Rendimiento , Concentración 50 Inhibidora , Luciferasas/metabolismo , Metabolómica , Parásitos/efectos de los fármacos , Parásitos/enzimología , Parásitos/crecimiento & desarrollo , Plasmodium falciparum/efectos de los fármacos , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/metabolismo , Sorbitol/farmacología
3.
Cell Microbiol ; 18(11): 1551-1569, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27019089

RESUMEN

The Plasmodium translocon for exported proteins (PTEX) has been established as the machinery responsible for the translocation of all classes of exported proteins beyond the parasitophorous vacuolar membrane of the intraerythrocytic malaria parasite. Protein export, particularly in the asexual blood stage, is crucial for parasite survival as exported proteins are involved in remodelling the host cell, an essential process for nutrient uptake, waste removal and immune evasion. Here, we have truncated the conserved C-terminus of one of the essential PTEX components, PTEX150, in Plasmodium falciparum in an attempt to create mutants of reduced functionality. Parasites tolerated C-terminal truncations of up to 125 amino acids with no reduction in growth, protein export or the establishment of new permeability pathways. Quantitative proteomic approaches however revealed a decrease in other PTEX subunits associating with PTEX150 in truncation mutants, suggesting a role for the C-terminus of PTEX150 in regulating PTEX stability. Our analyses also reveal three previously unreported PTEX-associated proteins, namely PV1, Pf113 and Hsp70-x (respective PlasmoDB numbers; PF3D7_1129100, PF3D7_1420700 and PF3D7_0831700) and demonstrate that core PTEX proteins exist in various distinct multimeric forms outside the major complex.


Asunto(s)
Eritrocitos/parasitología , Proteínas de Transporte de Membrana/fisiología , Plasmodium falciparum/fisiología , Proteoma/metabolismo , Proteínas Protozoarias/fisiología , Células Cultivadas , Humanos , Complejos Multiproteicos/metabolismo , Dominios Proteicos , Mapas de Interacción de Proteínas , Estabilidad Proteica , Transporte de Proteínas
4.
J Immunol ; 196(3): 1227-38, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26718341

RESUMEN

CXCL10, or IFN-γ-inducible protein 10, is a biomarker associated with increased risk for Plasmodium falciparum-mediated cerebral malaria (CM). Consistent with this, we have previously shown that CXCL10 neutralization or genetic deletion alleviates brain intravascular inflammation and protects Plasmodium berghei ANKA-infected mice from CM. In addition to organ-specific effects, the absence of CXCL10 during infection was also found to reduce parasite biomass. To identify the cellular sources of CXCL10 responsible for these processes, we irradiated and reconstituted wild-type (WT) and CXCL10(-/-) mice with bone marrow from either WT or CXCL10(-/-) mice. Similar to CXCL10(-/-) mice, chimeras unable to express CXCL10 in hematopoietic-derived cells controlled infection more efficiently than WT controls. In contrast, expression of CXCL10 in knockout mice reconstituted with WT bone marrow resulted in high parasite biomass levels, higher brain parasite and leukocyte sequestration rates, and increased susceptibility to CM. Neutrophils and inflammatory monocytes were identified as the main cellular sources of CXCL10 responsible for the induction of these processes. The improved control of parasitemia observed in the absence of CXCL10-mediated trafficking was associated with a preferential accumulation of CXCR3(+)CD4(+) T follicular helper cells in the spleen and enhanced Ab responses to infection. These results are consistent with the notion that some inflammatory responses elicited in response to malaria infection contribute to the development of high parasite densities involved in the induction of severe disease in target organs.


Asunto(s)
Quimiocina CXCL10/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Monocitos/inmunología , Neutrófilos/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Parasitemia/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
PLoS One ; 9(11): e112571, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25392998

RESUMEN

Drug discovery is a key part of malaria control and eradication strategies, and could benefit from sensitive and affordable assays to quantify parasite growth and to help identify the targets of potential anti-malarial compounds. Bioluminescence, achieved through expression of exogenous luciferases, is a powerful tool that has been applied in studies of several aspects of parasite biology and high throughput growth assays. We have expressed the new reporter NanoLuc (Nluc) luciferase in Plasmodium falciparum and showed it is at least 100 times brighter than the commonly used firefly luciferase. Nluc brightness was explored as a means to achieve a growth assay with higher sensitivity and lower cost. In addition we attempted to develop other screening assays that may help interrogate libraries of inhibitory compounds for their mechanism of action. To this end parasites were engineered to express Nluc in the cytoplasm, the parasitophorous vacuole that surrounds the intraerythrocytic parasite or exported to the red blood cell cytosol. As proof-of-concept, these parasites were used to develop functional screening assays for quantifying the effects of Brefeldin A, an inhibitor of protein secretion, and Furosemide, an inhibitor of new permeation pathways used by parasites to acquire plasma nutrients.


Asunto(s)
Proteínas Portadoras/genética , Ensayos Analíticos de Alto Rendimiento , Luciferasas/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Trofozoítos/crecimiento & desarrollo , Animales , Transporte Biológico , Brefeldino A/farmacología , Proteínas Portadoras/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/parasitología , Luciérnagas/química , Luciérnagas/enzimología , Furosemida/farmacología , Genes Reporteros , Humanos , Luciferasas/metabolismo , Penaeidae/química , Penaeidae/enzimología , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/metabolismo , Ingeniería de Proteínas , Inhibidores de la Síntesis de la Proteína/farmacología , Proteínas Protozoarias/metabolismo , Sensibilidad y Especificidad , Inhibidores del Simportador de Cloruro Sódico y Cloruro Potásico/farmacología , Trofozoítos/efectos de los fármacos , Trofozoítos/metabolismo
6.
Nature ; 511(7511): 587-91, 2014 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-25043043

RESUMEN

During the blood stages of malaria, several hundred parasite-encoded proteins are exported beyond the double-membrane barrier that separates the parasite from the host cell cytosol. These proteins have a variety of roles that are essential to virulence or parasite growth. There is keen interest in understanding how proteins are exported and whether common machineries are involved in trafficking the different classes of exported proteins. One potential trafficking machine is a protein complex known as the Plasmodium translocon of exported proteins (PTEX). Although PTEX has been linked to the export of one class of exported proteins, there has been no direct evidence for its role and scope in protein translocation. Here we show, through the generation of two parasite lines defective for essential PTEX components (HSP101 or PTEX150), and analysis of a line lacking the non-essential component TRX2 (ref. 12), greatly reduced trafficking of all classes of exported proteins beyond the double membrane barrier enveloping the parasite. This includes proteins containing the PEXEL motif (RxLxE/Q/D) and PEXEL-negative exported proteins (PNEPs). Moreover, the export of proteins destined for expression on the infected erythrocyte surface, including the major virulence factor PfEMP1 in Plasmodium falciparum, was significantly reduced in PTEX knockdown parasites. PTEX function was also essential for blood-stage growth, because even a modest knockdown of PTEX components had a strong effect on the parasite's capacity to complete the erythrocytic cycle both in vitro and in vivo. Hence, as the only known nexus for protein export in Plasmodium parasites, and an essential enzymic machine, PTEX is a prime drug target.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Malaria/parasitología , Plasmodium falciparum/genética , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Animales , Eritrocitos/metabolismo , Eritrocitos/parasitología , Proteínas de Choque Térmico/genética , Humanos , Estadios del Ciclo de Vida/fisiología , Complejos Multiproteicos/metabolismo , Transporte de Proteínas/genética , Proteínas Protozoarias/genética , Vacuolas/metabolismo , Vacuolas/parasitología
7.
PLoS Pathog ; 10(5): e1004135, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24854165

RESUMEN

To follow the fate of CD8+ T cells responsive to Plasmodium berghei ANKA (PbA) infection, we generated an MHC I-restricted TCR transgenic mouse line against this pathogen. T cells from this line, termed PbT-I T cells, were able to respond to blood-stage infection by PbA and two other rodent malaria species, P. yoelii XNL and P. chabaudi AS. These PbT-I T cells were also able to respond to sporozoites and to protect mice from liver-stage infection. Examination of the requirements for priming after intravenous administration of irradiated sporozoites, an effective vaccination approach, showed that the spleen rather than the liver was the main site of priming and that responses depended on CD8α+ dendritic cells. Importantly, sequential exposure to irradiated sporozoites followed two days later by blood-stage infection led to augmented PbT-I T cell expansion. These findings indicate that PbT-I T cells are a highly versatile tool for studying multiple stages and species of rodent malaria and suggest that cross-stage reactive CD8+ T cells may be utilized in liver-stage vaccine design to enable boosting by blood-stage infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunización Secundaria/métodos , Estadios del Ciclo de Vida/inmunología , Malaria/prevención & control , Plasmodium berghei/inmunología , Receptores de Antígenos de Linfocitos T/genética , Esporozoítos/inmunología , Traslado Adoptivo , Animales , Anopheles , Sangre/parasitología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Células Cultivadas , Hígado/inmunología , Hígado/parasitología , Malaria/sangre , Malaria/inmunología , Malaria/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasmodium berghei/crecimiento & desarrollo , Plasmodium chabaudi , Plasmodium yoelii , Receptores de Antígenos de Linfocitos T/inmunología
8.
PLoS One ; 9(4): e93268, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24691125

RESUMEN

BACKGROUND: The Natural Killer Complex (NKC) is a genetic region of highly linked genes encoding several receptors involved in the control of NK cell function. The NKC is highly polymorphic and allelic variability of various NKC loci has been demonstrated in inbred mice, providing evidence for NKC haplotypes. Using BALB.B6-Cmv1r congenic mice, in which NKC genes from C57BL/6 mice were introduced into the BALB/c background, we have previously shown that the NKC is a genetic determinant of malarial pathogenesis. C57BL/6 alleles are associated with increased disease-susceptibility as BALB.B6-Cmv1r congenic mice had increased cerebral pathology and death rates during P. berghei ANKA infection than cerebral malaria-resistant BALB/c controls. METHODS: To investigate which regions of the NKC are involved in susceptibility to experimental cerebral malaria (ECM), intra-NKC congenic mice generated by backcrossing recombinant F2 progeny from a (BALB/c x BALB.B6-Cmv1r) F1 intercross to BALB/c mice were infected with P. berghei ANKA. RESULTS: Our results revealed that C57BL/6 alleles at two locations in the NKC contribute to the development of ECM. The increased severity to severe disease in intra-NKC congenic mice was not associated with higher parasite burdens but correlated with a significantly enhanced systemic IFN-γ response to infection and an increased recruitment of CD8+ T cells to the brain of infected animals. CONCLUSIONS: Polymorphisms within the NKC modulate malarial pathogenesis and acquired immune responses to infection.


Asunto(s)
Sitios Genéticos , Lectinas Tipo C/genética , Malaria Cerebral/genética , Receptores de Superficie Celular/genética , Alelos , Animales , Biomarcadores , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Interferón gamma/biosíntesis , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos C57BL , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
9.
Parasitology ; 141(5): 602-13, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24476686

RESUMEN

Plasmodium falciparum malaria is responsible for over 250 million clinical cases every year worldwide. Severe malaria cases might present with a range of disease syndromes including acute respiratory distress, metabolic acidosis, hypoglycaemia, renal failure, anaemia, pulmonary oedema, cerebral malaria (CM) and placental malaria (PM) in pregnant women. Two main determinants of severe malaria have been identified: sequestration of parasitized red blood cells and strong pro-inflammatory responses. Increasing evidence from human studies and malaria infection animal models revealed the presence of host leucocytes at the site of parasite sequestration in brain blood vessels as well as placental tissue in complicated malaria cases. These observations suggested that apart from secreting cytokines, leucocytes might also contribute to disease by migrating to the site of parasite sequestration thereby exacerbating organ-specific inflammation. This evidence attracted substantial interest in identifying trafficking pathways by which inflammatory leucocytes are recruited to target organs during severe malaria syndromes. Chemo-attractant cytokines or chemokines are the key regulators of leucocyte trafficking and their potential contribution to disease has recently received considerable attention. This review summarizes the main findings to date, investigating the role of chemokines in severe malaria and the implication of these responses for the induction of pathogenesis and immunity to infection.


Asunto(s)
Quimiocinas/inmunología , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Receptores de Quimiocina/inmunología , Humanos , Malaria Falciparum/parasitología , Plasmodium falciparum/patogenicidad
10.
Biochem J ; 452(3): 433-41, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23548171

RESUMEN

PfCDPK1 [Plasmodium falciparum CDPK1 (calcium-dependent protein kinase 1)] is highly expressed in parasite asexual blood and mosquito stages. Its role is still poorly understood, but unsuccessful gene knockout attempts suggest that it is essential for parasite replication and/or RBC (red blood cell) invasion. In the present study, by tagging endogenous CDPK1 with GFP (green fluorescent protein), we demonstrate that CDPK1 localizes to the parasite plasma membrane of replicating and invasive forms as well as very young intracellular parasites and does not appear to be exported into RBCs. Although a knockdown of endogenous CDPK1 was achieved using a destabilization domain, parasites tolerated reduced expression without displaying a phenotype. Because of this, the PfCDPK1 auto-inhibitory J (junction) domain was explored as a means of achieving inducible and specific inhibition. Under in vitro conditions, a fusion protein comprising a J-GFP fusion specifically bound to PfCDPK1 and inhibited its activity. This fusion protein was conditionally expressed in P. falciparum asexual blood stages under the regulation of a DD (destabilization domain) (J-GFP-DD). We demonstrate that J-GFP-DD binds to CDPK1 and that this results in the arrest of parasite development late in the cell cycle during early schizogony. These data point to an early schizont function for PfCDPK1 and demonstrate that conditionally expressing auto-inhibitory regions can be an effective way to address the function of Plasmodium enzymes.


Asunto(s)
Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/metabolismo , Proteínas Quinasas/biosíntesis , Proteínas Protozoarias/antagonistas & inhibidores , Proteínas Protozoarias/biosíntesis , Esquizontes/crecimiento & desarrollo , Esquizontes/metabolismo , Células Cultivadas , Plasmodium falciparum/enzimología , Estructura Terciaria de Proteína/genética , Proteínas Protozoarias/genética , Esquizontes/enzimología
11.
Immunobiology ; 218(2): 263-71, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22704523

RESUMEN

Cerebral malaria (CM) is the most severe syndrome associated with Plasmodium falciparum infections. Experimental evidence suggests that disease results from the sequestration of parasitized-red blood cells (pRBCs) together with inflammatory leukocytes within brain capillaries. We have previously shown that NK cells stimulate migration of CXCR3(+) T cells to the brain of Plasmodium berghei ANKA-infected mice. Here we investigated whether interactions between NK cells and dendritic cells (DCs) are required for the induction of T cell responses involved in disease. For that, NK cell-depleted and control mice were infected with transgenic parasites expressing model T cell epitopes. T cells from TCR transgenic mice specific for those epitopes were adoptively transferred and proliferation was determined. NK cell depletion significantly reduced CD8(+) but not CD4(+) DC-mediated T cell priming. Lack of NK cells did not compromise CD8(+) T cell responses in IL-12(-/-) mice, suggesting that NK cells stimulate IL-12 output by DCs required for optimal T cell priming. The contribution of DCs to NK cell function was also investigated. DC depletion and genetic deletion of IL-12 dramatically reduced NK cell-mediated IFN-γ responses to malaria. Thus NK cells and DCs engage in reciprocal activation for the induction of inflammatory responses involved in severe malaria.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Malaria Cerebral/inmunología , Plasmodium berghei/inmunología , Traslado Adoptivo , Animales , Animales Modificados Genéticamente , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/trasplante , Comunicación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Epítopos de Linfocito T/genética , Humanos , Interferón gamma/metabolismo , Interleucina-12/genética , Activación de Linfocitos/genética , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasmodium berghei/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores CXCR3/metabolismo
12.
PLoS Pathog ; 5(4): e1000369, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19343215

RESUMEN

Plasmodium falciparum malaria causes 660 million clinical cases with over 2 million deaths each year. Acquired host immunity limits the clinical impact of malaria infection and provides protection against parasite replication. Experimental evidence indicates that cell-mediated immune responses also result in detrimental inflammation and contribute to severe disease induction. In both humans and mice, the spleen is a crucial organ involved in blood stage malaria clearance, while organ-specific disease appears to be associated with sequestration of parasitized erythrocytes in vascular beds and subsequent recruitment of inflammatory leukocytes. Using a rodent model of cerebral malaria, we have previously found that the majority of T lymphocytes in intravascular infiltrates of cerebral malaria-affected mice express the chemokine receptor CXCR3. Here we investigated the effect of IP-10 blockade in the development of experimental cerebral malaria and the induction of splenic anti-parasite immunity. We found that specific neutralization of IP-10 over the course of infection and genetic deletion of this chemokine in knockout mice reduces cerebral intravascular inflammation and is sufficient to protect P. berghei ANKA-infected mice from fatality. Furthermore, our results demonstrate that lack of IP-10 during infection significantly reduces peripheral parasitemia. The increased resistance to infection observed in the absence of IP-10-mediated cell trafficking was associated with retention and subsequent expansion of parasite-specific T cells in spleens of infected animals, which appears to be advantageous for the control of parasite burden. Thus, our results demonstrate that modulating homing of cellular immune responses to malaria is critical for reaching a balance between protective immunity and immunopathogenesis.


Asunto(s)
Quimiocina CXCL10/inmunología , Malaria Cerebral/inmunología , Malaria/inmunología , Plasmodium berghei/fisiología , Linfocitos T/inmunología , Animales , Química Encefálica , Quimiocina CXCL10/genética , Modelos Animales de Enfermedad , Inmunidad Celular , Inflamación , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pruebas de Neutralización , Parasitemia/metabolismo , Bazo/inmunología , Estadísticas no Paramétricas
13.
Proc Natl Acad Sci U S A ; 105(38): 14509-14, 2008 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-18799734

RESUMEN

Although CD8(+) T cells do not contribute to protection against the blood stage of Plasmodium infection, there is mounting evidence that they are principal mediators of murine experimental cerebral malaria (ECM). At present, there is no direct evidence that the CD8(+) T cells mediating ECM are parasite-specific or, for that matter, whether parasite-specific CD8(+) T cells are generated in response to blood-stage infection. To resolve this and to define the cellular requirements for such priming, we generated transgenic P. berghei parasites expressing model T cell epitopes. This approach was necessary as MHC class I-restricted antigens to blood-stage infection have not been defined. Here, we show that blood-stage infection leads to parasite-specific CD8(+) and CD4(+) T cell responses. Furthermore, we show that P. berghei-expressed antigens are cross-presented by the CD8alpha(+) subset of dendritic cells (DC), and that this induces pathogen-specific cytotoxic T lymphocytes (CTL) capable of lysing cells presenting antigens expressed by blood-stage parasites. Finally, using three different experimental approaches, we provide evidence that CTL specific for parasite-expressed antigens contribute to ECM.


Asunto(s)
Antígenos de Protozoos/inmunología , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Plasmodium berghei/inmunología , Animales , Animales Modificados Genéticamente , Encéfalo/inmunología , Citotoxicidad Inmunológica , Epítopos de Linfocito T/inmunología , Estadios del Ciclo de Vida , Malaria Cerebral/sangre , Malaria Cerebral/mortalidad , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo
14.
Eur J Immunol ; 38(4): 1082-95, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18383042

RESUMEN

Cerebral malaria (CM) results from the binding of infected erythrocytes and leukocytes to brain endothelia. The precise mechanisms underlying lymphocyte recruitment and activation in CM remain unclear. Therefore, the expression of various chemokines was quantified in brains of mice infected with Plasmodium berghei ANKA (PbA). Several chemokines attracting monocytes and activated T-lymphocytes were expressed at high levels. Their expression was almost completely abrogated in IFN-gamma ligand and receptor KO mice, indicating that IFN-gamma is an essential chemokine inducer in vivo. Surprisingly, the expression levels of chemokines, IFN-gamma and also adhesion molecules in the brain were not lower in CM-resistant Balb/c and DBA/2 mice compared to CM-sensitive C57BL/6 and DBA/1 mice, although T lymphocyte sequestration in the brain was significantly less in CM-resistant than in CM-sensitive mice. This difference correlated with a higher up-regulation of the CXC chemokine receptor (CXCR)-3 on splenic T cells and a higher chemotactic response to IFN-gamma-inducible protein-10 (IP-10) in C57BL/6 compared to Balb/c mice. In conclusion, parasite-induced IFN-gamma in the brain results in high local expression levels of specific chemokines for monocytes and lymphocytes. The strain-dependent susceptibility to develop CM is more related to the expression of CXCR3 in circulating leukocytes than to the chemokine expression levels in the brain.


Asunto(s)
Movimiento Celular/inmunología , Quimiocinas/inmunología , Interferón gamma/inmunología , Malaria Cerebral/inmunología , Receptores CXCR3/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Animales , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Quimiocina CXCL6/genética , Quimiocina CXCL6/inmunología , Quimiocinas/genética , Quimiocinas/metabolismo , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Ligandos , Macrófagos/citología , Macrófagos/inmunología , Malaria Cerebral/parasitología , Ratones , Monocitos/citología , Monocitos/inmunología , Plasmodium berghei/inmunología , Receptores de Interferón/metabolismo , Bazo/citología , Bazo/inmunología , Tasa de Supervivencia , Receptor de Interferón gamma
15.
J Immunol ; 178(9): 5779-88, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17442962

RESUMEN

NK cells are cytotoxic lymphocytes that also secrete regulatory cytokines and can therefore influence adaptive immune responses. NK cell function is largely controlled by genes present in a genomic region named the NK complex. It has been shown that the NK complex is a genetic determinant of murine cerebral malaria pathogenesis mediated by Plasmodium berghei ANKA. In this study, we show that NK cells are required for cerebral malaria disease induction and the control of parasitemia. NK cells were found infiltrating brains of cerebral malaria-affected mice. NK cell depletion resulted in inhibition of T cell recruitment to the brain of P. berghei-infected animals. NK cell-depleted mice displayed down-regulation of CXCR3 expression and a significant reduction of T cells migrating in response to IFN-gamma-inducible protein 10, indicating that this chemokine pathway plays an essential role in leukocyte trafficking leading to cerebral disease and fatalities.


Asunto(s)
Encéfalo/inmunología , Células Asesinas Naturales/inmunología , Malaria Cerebral/inmunología , Plasmodium berghei , Receptores de Quimiocina/metabolismo , Subgrupos de Linfocitos T/inmunología , Animales , Encéfalo/patología , Quimiocina CXCL10 , Quimiocinas CX3C/metabolismo , Quimiocinas CX3C/farmacología , Quimiocinas CXC/metabolismo , Quimiocinas CXC/farmacología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Depleción Linfocítica , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Receptores CXCR3 , Receptores de Quimiocina/análisis , Bazo/inmunología , Subgrupos de Linfocitos T/química , Subgrupos de Linfocitos T/efectos de los fármacos
16.
Infect Immun ; 75(5): 2275-82, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17325053

RESUMEN

The infection of mice with Plasmodium berghei ANKA constitutes the best available mouse model for human Plasmodium falciparum-mediated cerebral malaria, a devastating neurological syndrome that kills nearly 2.5 million people every year. Experimental data suggest that cerebral disease results from the sequestration of parasitized erythrocytes within brain blood vessels, which is exacerbated by host proinflammatory responses mediated by cytokines and effector cells including T lymphocytes. Here, T cell responses to P. berghei ANKA were analyzed in cerebral malaria-resistant and -susceptible mouse strains. CD4+ T-cell proliferation and interleukin-2 (IL-2) production in response to parasite-specific and polyclonal stimuli were strongly inhibited in cerebral malaria-resistant mice. In vitro and in vivo depletion of CD4+ CD25+ regulatory T (T(reg)) cells significantly reversed the inhibition of CD4+ T-cell proliferation and IL-2 production, indicating that this cell population contributes to the suppression of T-cell function during malaria. Moreover, in vivo depletion of T(reg) cells prevented the development of parasite-specific TH1 cells involved in the induction of cerebral malaria during a secondary parasitic challenge, demonstrating a regulatory role for this cell population in the control of pathogenic responses leading to fatal disease.


Asunto(s)
Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Plasmodium berghei/patogenicidad , Linfocitos T Reguladores/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Activación de Linfocitos , Malaria Cerebral/inmunología , Malaria Cerebral/mortalidad , Malaria Cerebral/parasitología , Malaria Cerebral/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células TH1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...