Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
PLoS Pathog ; 8(10): e1002982, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133375

RESUMEN

Plasmodium falciparum malaria is responsible for nearly one million annual deaths worldwide. Because of the difficulty in monitoring the pathogenesis of cerebral malaria in humans, we conducted a study in various mouse models to better understand disease progression in experimental cerebral malaria (ECM). We compared the effect on the integrity of the blood brain barrier (BBB) and the histopathology of the brain of P. berghei ANKA, a known ECM model, P. berghei NK65, generally thought not to induce ECM, P. yoelii 17XL, originally reported to induce human cerebral malaria-like histopathology, and P. yoelii YM. As expected, P. berghei ANKA infection caused neurological signs, cerebral hemorrhages, and BBB dysfunction in CBA/CaJ and Swiss Webster mice, while Balb/c and A/J mice were resistant. Surprisingly, PbNK induced ECM in CBA/CaJ mice, while all other mice were resistant. P. yoelii 17XL and P. yoelii YM caused lethal hyperparasitemia in all mouse strains; histopathological alterations, BBB dysfunction, or neurological signs were not observed. Intravital imaging revealed that infected erythrocytes containing mature parasites passed slowly through capillaries making intimate contact with the endothelium, but did not arrest. Except for relatively rare microhemorrhages, mice with ECM presented no obvious histopathological alterations that would explain the widespread disruption of the BBB. Intravital imaging did reveal, however, that postcapillary venules, but not capillaries or arterioles, from mice with ECM, but not hyperparasitemia, exhibit platelet marginalization, extravascular fibrin deposition, CD14 expression, and extensive vascular leakage. Blockage of LFA-1 mediated cellular interactions prevented leukocyte adhesion, vascular leakage, neurological signs, and death from ECM. The endothelial barrier-stabilizing mediators imatinib and FTY720 inhibited vascular leakage and neurological signs and prolonged survival to ECM. Thus, it appears that neurological signs and coma in ECM are due to regulated opening of paracellular-junctional and transcellular-vesicular fluid transport pathways at the neuroimmunological BBB.


Asunto(s)
Barrera Hematoencefálica/patología , Malaria Cerebral/patología , Plasmodium berghei/patogenicidad , Plasmodium yoelii/patogenicidad , Animales , Benzamidas , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/fisiopatología , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/etiología , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod , Mesilato de Imatinib , Receptores de Lipopolisacáridos/biosíntesis , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Malaria Cerebral/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos CBA , Neuroinmunomodulación , Parasitemia , Piperazinas/farmacología , Plasmodium falciparum/patogenicidad , Glicoles de Propileno/farmacología , Pirimidinas/farmacología , Esfingosina/análogos & derivados , Esfingosina/farmacología
2.
PLoS Pathog ; 3(11): e171, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17997605

RESUMEN

Plasmodium undergoes one round of multiplication in the liver prior to invading erythrocytes and initiating the symptomatic blood phase of the malaria infection. Productive hepatocyte infection by sporozoites leads to the generation of thousands of merozoites capable of erythrocyte invasion. Merozoites are released from infected hepatocytes as merosomes, packets of hundreds of parasites surrounded by host cell membrane. Intravital microscopy of green fluorescent protein-expressing P. yoelii parasites showed that the majority of merosomes exit the liver intact, adapt a relatively uniform size of 12-18 microm, and contain 100-200 merozoites. Merosomes survived the subsequent passage through the right heart undamaged and accumulated in the lungs. Merosomes were absent from blood harvested from the left ventricle and from tail vein blood, indicating that the lungs effectively cleared the blood from all large parasite aggregates. Accordingly, merosomes were not detectable in major organs such as brain, kidney, and spleen. The failure of annexin V to label merosomes collected from hepatic effluent indicates that phosphatidylserine is not exposed on the surface of the merosome membrane suggesting the infected hepatocyte did not undergo apoptosis prior to merosome release. Merosomal merozoites continued to express green fluorescent protein and did not incorporate propidium iodide or YO-PRO-1 indicating parasite viability and an intact merosome membrane. Evidence of merosomal merozoite infectivity was provided by hepatic effluent containing merosomes being significantly more infective than blood with an identical low-level parasitemia. Ex vivo analysis showed that merosomes eventually disintegrate inside pulmonary capillaries, thus liberating merozoites into the bloodstream. We conclude that merosome packaging protects hepatic merozoites from phagocytic attack by sinusoidal Kupffer cells, and that release into the lung microvasculature enhances the chance of successful erythrocyte invasion. We believe this previously unknown part of the plasmodial life cycle ensures an effective transition from the liver to the blood phase of the malaria infection.


Asunto(s)
Hígado/parasitología , Malaria/parasitología , Merozoítos/fisiología , Plasmodium yoelii/fisiología , Circulación Pulmonar , Animales , Pulmón/irrigación sanguínea , Pulmón/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Microscopía Confocal
3.
Subcell Biochem ; 47: 182-97, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18512352

RESUMEN

Malaria sporozoites must cross at least two cell barriers to reach their initial site of replication in the mammalian host. After transmission into the skin by an infected mosquito, they migrate towards small dermal capillaries, traverse the vascular endothelial layer, and rapidly home to the liver. To infect hepatocytes, the parasites must cross the sinusoidal cell layer, composed of specialized highly fenestrated sinusoidal endothelia and Kupffer cells, the resident macrophages of the liver (Fig. 1). The exact route Plasmodium sporozoites take to hepatocytes has been subject of controversial discussions for many years. Recent cell biological, microscopic, and genetic approaches have considerably enhanced our understanding of the initial events leading to the establishment of a malaria infection in the liver.


Asunto(s)
Hígado/parasitología , Plasmodium/patogenicidad , Esporozoítos/fisiología , Animales , Hepatocitos/inmunología , Hepatocitos/parasitología , Humanos , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/parasitología , Malaria/inmunología , Malaria/parasitología , Modelos Biológicos , Plasmodium/inmunología , Plasmodium/fisiología , Esporozoítos/inmunología
4.
Int J Parasitol ; 36(12): 1283-93, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16890231

RESUMEN

The liver stages of Plasmodium, the causative agent of malaria, are the least explored forms in the parasite's life cycle despite their recognition as key vaccine and drug targets. In vivo experimental access to liver stages of human malaria parasites is practically prohibited and therefore rodent model malaria parasites have been used for in vivo studies. However, even in rodent models progress in the analysis of liver stages has been limited, mainly due to their low abundance and associated difficulties in visualisation and isolation. Here, we present green fluorescent protein (GFP)-tagged Plasmodium yoelii rodent malaria parasite liver infections in BALB/c mice as an excellent quantitative model for the live visualisation and isolation of the so far elusive liver stages. We believe P. yoelii GFP-tagged liver stages allow, for the first time, the efficient quantitative isolation of intact early and late liver stage-infected hepatocyte units by fluorescence activated cell sorting. GFP-tagged liver stages are also well suited for intravital imaging, allowing us for the first time to visualise them in real time. We identify previously unrecognised features of liver stages including vigorous parasite movement and expulsion of 'extrusomes'. Intravital imaging thus reveals new, important information on the malaria parasite's transition from tissue to blood stage.


Asunto(s)
Parasitosis Hepáticas/parasitología , Malaria/parasitología , Plasmodium yoelii/aislamiento & purificación , Animales , Animales Modificados Genéticamente , Línea Celular , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo/métodos , Genotipo , Proteínas Fluorescentes Verdes , Hepatocitos/parasitología , Hígado/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Microscopía Fluorescente/métodos , Fenotipo , Plasmodium berghei/genética , Plasmodium berghei/aislamiento & purificación , Plasmodium yoelii/genética , Transfección/métodos
5.
Cell Microbiol ; 9(2): 397-412, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16953803

RESUMEN

Previous studies suggested Plasmodium sporozoites infect hepatocytes after passing through Kupffer cells, but proof has been elusive. Here we present new information strengthening that hypothesis. We used homozygous op/op mice known to have few Kupffer cells because they lack macrophage colony stimulating factor 1 required for macrophage maturation due to a deactivating point mutation in the osteopetrosis gene. We found these mice to have 77% fewer Kupffer cells and to exhibit reduced clearance of colloidal carbon particles compared with heterozygous phenotypically normal littermates. Using a novel quantitative reverse transcription polymerase chain reaction assay for P. yoelii 18S rRNA, we found liver infection of op/op mice to be decreased by 84% compared with controls. However, using another way of limiting Kupffer cells, treatment with liposome-encapsulated clodronate, infection of normal mice was enhanced seven- to 15-fold. This was explained by electron microscopy showing temporary gaps in the sinusoidal cell layer caused by this treatment. Thus, Kupffer cell deficiency in op/op mice decreases sporozoite infection by reducing the number of portals to the liver parenchyma, whereas clodronate increases sporozoite infection by opening portals and providing direct access to hepatocytes. Together these data provide strong support for the hypothesis that Kupffer cells are the portal for sporozoites to hepatocytes and critical for the onset of a malaria infection.


Asunto(s)
Macrófagos del Hígado/fisiología , Hígado/parasitología , Plasmodium yoelii/patogenicidad , Proteínas Protozoarias/fisiología , Esporozoítos/fisiología , Animales , Macrófagos del Hígado/parasitología , Hígado/citología , Hígado/metabolismo , Ratones , Plasmodium yoelii/crecimiento & desarrollo , Proteínas Protozoarias/biosíntesis
6.
Cell Microbiol ; 8(10): 1537-46, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16911567

RESUMEN

The initial site of replication for Plasmodium parasites in mammalian hosts are hepatocytes, cells that offer unique advantages for the extensive parasite replication occurring prior to the erythrocytic phase of the life cycle. The liver is the metabolic centre of the body and has an unusual relationship to the immune system. However, to reach hepatocytes, sporozoites must cross the sinusoidal barrier, composed of specialized endothelia and Kupffer cells, the resident macrophages of the liver. Mounting evidence suggests that, instead of taking what would seem a safer route through endothelia, the parasites traverse Kupffer cells yet suffer no harm. Kupffer cells have a broad range of responses towards incoming microorganisms, toxins and antigens which depend on the nature of the intruder, the experimental conditions and the environmental circumstances. Kupffer cells may become activated or remain anergic, produce pro- or anti-inflammatory mediators. Consequently, outcomes are diverse and include development of immunity or tolerance, parenchymal necrosis or regeneration, chronic cirrhotic transformation or acute liver failure. Here we review data concerning the unique structural and functional characteristics of Kupffer cells and their interactions with Plasmodium sporozoites in the context of a model in which these hepatic macrophages function as the sporozoite gate to the liver.


Asunto(s)
Macrófagos del Hígado/parasitología , Malaria/parasitología , Plasmodium/fisiología , Esporozoítos/fisiología , Animales , Humanos , Tolerancia Inmunológica , Macrófagos del Hígado/citología , Macrófagos del Hígado/inmunología , Estadios del Ciclo de Vida , Hígado/citología , Hígado/inmunología , Hígado/parasitología , Malaria/inmunología , Plasmodium/inmunología , Plasmodium/metabolismo , Vena Porta/inmunología , Vena Porta/parasitología , Esporozoítos/inmunología , Esporozoítos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA