Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(26): e2204581119, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35727988

RESUMEN

The brain microcirculation is increasingly viewed as a potential target for disease-modifying drugs in the treatment of Alzheimer's disease patients, reflecting a growing appreciation of evidence that cerebral blood flow is compromised in such patients. However, the pathogenic mechanisms in brain resistance arteries underlying blood flow defects have not yet been elucidated. Here we probed the roles of principal vasodilatory pathways in cerebral arteries using the APP23 mouse model of Alzheimer's disease, in which amyloid precursor protein is increased approximately sevenfold, leading to neuritic plaques and cerebrovascular accumulation of amyloid-ß similar to those in patients with Alzheimer's disease. Pial arteries from APP23 mice (18 mo old) exhibited enhanced pressure-induced (myogenic) constriction because of a profound reduction in ryanodine receptor-mediated, local calcium-release events ("Ca2+ sparks") in arterial smooth muscle cells and a consequent decrease in the activity of large-conductance Ca2+-activated K+ (BK) channels. The ability of the endothelial cell inward rectifier K+ (Kir2.1) channel to cause dilation was also compromised. Acute application of amyloid-ß 1-40 peptide to cerebral arteries from wild-type mice partially recapitulated the BK dysfunction seen in APP23 mice but had no effect on Kir2.1 function. If mirrored in human Alzheimer's disease, these tandem defects in K+ channel-mediated vasodilation could account for the clinical cerebrovascular presentation seen in patients: reduced blood flow and crippled functional hyperemia. These data direct future research toward approaches that reverse this dual vascular channel dysfunction, with the ultimate aim of restoring healthy cerebral blood flow and improving clinical outcomes.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , Señalización del Calcio , Canales de Potasio de Gran Conductancia Activados por el Calcio , Músculo Liso Vascular , Miocitos del Músculo Liso , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Encéfalo/irrigación sanguínea , Arterias Cerebrales/metabolismo , Modelos Animales de Enfermedad , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Vasodilatación
2.
Immunology ; 164(4): 737-753, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34407221

RESUMEN

Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterized by CD8+ T-cell accumulation within the brain. Whilst the dynamics of CD8+ T-cell activation and migration during extant primary PbA infection have been extensively researched, the fate of the parasite-specific CD8+ T cells upon resolution of ECM is not understood. In this study, we show that memory OT-I cells persist systemically within the spleen, lung and brain following recovery from ECM after primary PbA-OVA infection. Whereas memory OT-I cells within the spleen and lung exhibited canonical central memory (Tcm) and effector memory (Tem) phenotypes, respectively, memory OT-I cells within the brain post-PbA-OVA infection displayed an enriched CD69+ CD103- profile and expressed low levels of T-bet. OT-I cells within the brain were excluded from short-term intravascular antibody labelling but were targeted effectively by longer-term systemically administered antibodies. Thus, the memory OT-I cells were extravascular within the brain post-ECM but were potentially not resident memory cells. Importantly, whilst memory OT-I cells exhibited strong reactivation during secondary PbA-OVA infection, preventing activation of new primary effector T cells, they had dampened reactivation during a fourth PbA-OVA infection. Overall, our results demonstrate that memory CD8+ T cells are systemically distributed but exhibit a unique phenotype within the brain post-ECM, and that their reactivation characteristics are shaped by infection history. Our results raise important questions regarding the role of distinct memory CD8+ T-cell populations within the brain and other tissues during repeat Plasmodium infections.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interacciones Huésped-Parásitos/inmunología , Malaria/inmunología , Malaria/parasitología , Plasmodium berghei/fisiología , Animales , Biomarcadores , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Quimiotaxis de Leucocito/inmunología , Susceptibilidad a Enfermedades , Epítopos de Linfocito T/inmunología , Eritrocitos/inmunología , Eritrocitos/parasitología , Matriz Extracelular , Memoria Inmunológica , Inmunofenotipificación , Estadios del Ciclo de Vida , Activación de Linfocitos/inmunología , Malaria/metabolismo , Malaria/patología , Malaria Cerebral/inmunología , Malaria Cerebral/metabolismo , Malaria Cerebral/parasitología , Ratones , Ratones Transgénicos , Especificidad de Órganos/inmunología
3.
J Cereb Blood Flow Metab ; 40(7): 1402-1414, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32151228

RESUMEN

Assessment of outcome in preclinical studies of vascular cognitive impairment (VCI) is heterogenous. Through an ARUK Scottish Network supported questionnaire and workshop (mostly UK-based researchers), we aimed to determine underlying variability and what could be implemented to overcome identified challenges. Twelve UK VCI research centres were identified and invited to complete a questionnaire and attend a one-day workshop. Questionnaire responses demonstrated agreement that outcome assessments in VCI preclinical research vary by group and even those common across groups, may be performed differently. From the workshop, six themes were discussed: issues with preclinical models, reasons for choosing functional assessments, issues in interpretation of functional assessments, describing and reporting functional outcome assessments, sharing resources and expertise, and standardization of outcomes. Eight consensus points emerged demonstrating broadly that the chosen assessment should reflect the deficit being measured, and therefore that one assessment does not suit all models; guidance/standardisation on recording VCI outcome reporting is needed and that uniformity would be aided by a platform to share expertise, material, protocols and procedures thus reducing heterogeneity and so increasing potential for collaboration, comparison and replication. As a result of the workshop, UK wide consensus statements were agreed and future priorities for preclinical research identified.


Asunto(s)
Demencia Vascular , Modelos Animales de Enfermedad , Proyectos de Investigación/normas , Animales , Consenso , Recuperación de la Función , Encuestas y Cuestionarios , Reino Unido
4.
Front Immunol ; 10: 1747, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396236

RESUMEN

CD8+ T cells have been shown to play a critical role in the pathogenesis of experimental cerebral malaria (ECM) in mice, but their role in development of human cerebral malaria (HCM) remains unclear. Thus, in this study we have provided the first direct contrast of the accumulation of CD8+ T cells in the brain during HCM and ECM. HCM cases were from children who died of Plasmodium falciparum cerebral malaria at Queen Elizabeth Central Hospital (Malawi) between 2003 and 2010. ECM was induced by infecting C57BL/6J mice with P. berghei ANKA. We demonstrate similarities in the intracerebral CD8+ T cell responses in ECM and HCM, in particular an apparent shared choroid plexus-meningeal route of CD8+ T cell accumulation in the brain. Nevertheless, we also reveal some potentially important differences in compartmentalization of CD8+ T cells within the cerebrovascular bed in HCM and ECM.


Asunto(s)
Encéfalo , Linfocitos T CD8-positivos , Malaria Cerebral , Malaria Falciparum , Plasmodium berghei/inmunología , Plasmodium falciparum/inmunología , Animales , Encéfalo/inmunología , Encéfalo/parasitología , Encéfalo/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Niño , Preescolar , Femenino , Humanos , Malaria Cerebral/inmunología , Malaria Cerebral/patología , Malaria Falciparum/inmunología , Malaria Falciparum/patología , Masculino , Ratones
5.
Front Immunol ; 10: 248, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30846985

RESUMEN

Cerebral malaria (CM) is one of the most severe complications of Plasmodium falciparum infection. There is evidence that repeated parasite exposure promotes resistance against CM. However, the immunological basis of this infection-induced resistance remains poorly understood. Here, utilizing the Plasmodium berghei ANKA (PbA) model of experimental cerebral malaria (ECM), we show that three rounds of infection and drug-cure protects against the development of ECM during a subsequent fourth (4X) infection. Exposure-induced resistance was associated with specific suppression of CD8+ T cell activation and CTL-related pathways, which corresponded with the development of heterogeneous atypical B cell populations as well as the gradual infection-induced generation and maintenance of high levels of anti-parasite IgG. Mechanistically, transfer of high-titer anti-parasite IgG did not protect 1X infected mice against ECM and depletion of atypical and regulatory B cells during 4X infection failed to abrogate infection-induced resistance to ECM. However, IgMi mice that were unable to produce secreted antibody, or undergo class switching, during the repeated rounds of infection failed to develop resistance against ECM. The failure of infection-induced protection in IgMi mice was associated with impaired development of atypical B cell populations and the inability to suppress pathogenic CD8+ T cell responses. Our results, therefore, suggest the importance of anti-parasite antibody responses, gradually acquired, and maintained through repeated Plasmodium infections, for modulating the B cell compartment and eventually suppressing memory CD8+ T cell reactivation to establish infection-induced resistance to ECM.


Asunto(s)
Formación de Anticuerpos/inmunología , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Malaria Cerebral/inmunología , Plasmodium berghei/inmunología , Animales , Encéfalo/parasitología , Linfocitos T CD8-positivos/parasitología , Activación de Linfocitos/inmunología , Malaria Cerebral/parasitología , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Ratones , Ratones Endogámicos C57BL
6.
Proc Natl Acad Sci U S A ; 115(28): 7404-7409, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29954866

RESUMEN

Cerebral malaria (CM) is a serious neurological complication caused by Plasmodium falciparum infection. Currently, the only treatment for CM is the provision of antimalarial drugs; however, such treatment by itself often fails to prevent death or development of neurological sequelae. To identify potential improved treatments for CM, we performed a nonbiased whole-brain transcriptomic time-course analysis of antimalarial drug chemotherapy of murine experimental CM (ECM). Bioinformatics analyses revealed IL33 as a critical regulator of neuroinflammation and cerebral pathology that is down-regulated in the brain during fatal ECM and in the acute period following treatment of ECM. Consistent with this, administration of IL33 alongside antimalarial drugs significantly improved the treatment success of established ECM. Mechanistically, IL33 treatment reduced inflammasome activation and IL1ß production in microglia and intracerebral monocytes in the acute recovery period following treatment of ECM. Moreover, treatment with the NLRP3-inflammasome inhibitor MCC950 alongside antimalarial drugs phenocopied the protective effect of IL33 therapy in improving the recovery from established ECM. We further showed that IL1ß release from macrophages was stimulated by hemozoin and antimalarial drugs and that this was inhibited by MCC950. Our results therefore demonstrate that manipulation of the IL33-NLRP3 axis may be an effective therapy to suppress neuroinflammation and improve the efficacy of antimalarial drug treatment of CM.


Asunto(s)
Antimaláricos/farmacología , Encéfalo/parasitología , Sistemas de Liberación de Medicamentos/métodos , Interleucina-33/metabolismo , Malaria Cerebral/tratamiento farmacológico , Malaria Falciparum/tratamiento farmacológico , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Plasmodium falciparum/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Hemoproteínas/metabolismo , Interleucina-1beta/biosíntesis , Interleucina-33/antagonistas & inhibidores , Macrófagos/metabolismo , Macrófagos/patología , Malaria Cerebral/metabolismo , Malaria Cerebral/patología , Malaria Falciparum/metabolismo , Malaria Falciparum/patología , Masculino , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Transcriptoma/efectos de los fármacos
7.
J Exp Med ; 215(6): 1507-1518, 2018 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-29789388

RESUMEN

A defining feature of resident gut macrophages is their high replenishment rate from blood monocytes attributed to tonic commensal stimulation of this site. In contrast, almost all other tissues contain locally maintained macrophage populations, which coexist with monocyte-replenished cells at homeostasis. In this study, we identified three transcriptionally distinct mouse gut macrophage subsets that segregate based on expression of Tim-4 and CD4. Challenging current understanding, Tim-4+CD4+ gut macrophages were found to be locally maintained, while Tim-4-CD4+ macrophages had a slow turnover from blood monocytes; indeed, Tim-4-CD4- macrophages were the only subset with the high monocyte-replenishment rate currently attributed to gut macrophages. Moreover, all macrophage subpopulations required live microbiota to sustain their numbers, not only those derived from blood monocytes. These findings oppose the prevailing paradigm that all macrophages in the adult mouse gut rapidly turn over from monocytes in a microbiome-dependent manner; instead, these findings supplant it with a model of ontogenetic diversity where locally maintained subsets coexist with rapidly replaced monocyte-derived populations.


Asunto(s)
Antígenos CD4/metabolismo , Intestinos/citología , Macrófagos/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Animales Recién Nacidos , Intestinos/microbiología , Ratones Endogámicos C57BL , Microbiota , Monocitos/metabolismo , Fenotipo , Receptores CCR2/metabolismo , Transcripción Genética
8.
Infect Immun ; 85(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28874445

RESUMEN

Experimental cerebral malaria (ECM) is a gamma interferon (IFN-γ)-dependent syndrome. However, whether IFN-γ promotes ECM through direct and synergistic targeting of multiple cell populations or by acting primarily on a specific responsive cell type is currently unknown. Here, using a panel of cell- and compartment-specific IFN-γ receptor 2 (IFN-γR2)-deficient mice, we show that IFN-γ causes ECM by signaling within both the hematopoietic and nonhematopoietic compartments. Mechanistically, hematopoietic and nonhematopoietic compartment-specific IFN-γR signaling exerts additive effects in orchestrating intracerebral inflammation, leading to the development of ECM. Surprisingly, mice with specific deletion of IFN-γR2 expression on myeloid cells, T cells, or neurons were completely susceptible to terminal ECM. Utilizing a reductionist in vitro system, we show that synergistic IFN-γ and tumor necrosis factor (TNF) stimulation promotes strong activation of brain blood vessel endothelial cells. Combined, our data show that within the hematopoietic compartment, IFN-γ causes ECM by acting redundantly or by targeting non-T cell or non-myeloid cell populations. Within the nonhematopoietic compartment, brain endothelial cells, but not neurons, may be the major target of IFN-γ leading to ECM development. Collectively, our data provide information on how IFN-γ mediates the development of cerebral pathology during malaria infection.


Asunto(s)
Encéfalo/inmunología , Células Endoteliales/inmunología , Interferón gamma/genética , Malaria Cerebral/genética , Plasmodium berghei/patogenicidad , Receptores de Interferón/genética , Animales , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Encéfalo/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Modelos Animales de Enfermedad , Células Endoteliales/parasitología , Regulación de la Expresión Génica , Interferón gamma/inmunología , Interleucinas/genética , Interleucinas/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/parasitología , Neuronas/inmunología , Neuronas/parasitología , Plasmodium berghei/inmunología , Receptores de Interferón/deficiencia , Receptores de Interferón/inmunología , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/parasitología
9.
PLoS Pathog ; 13(3): e1006267, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28273147

RESUMEN

The murine model of experimental cerebral malaria (ECM) has been utilised extensively in recent years to study the pathogenesis of human cerebral malaria (HCM). However, it has been proposed that the aetiologies of ECM and HCM are distinct, and, consequently, no useful mechanistic insights into the pathogenesis of HCM can be obtained from studying the ECM model. Therefore, in order to determine the similarities and differences in the pathology of ECM and HCM, we have performed the first spatial and quantitative histopathological assessment of the ECM syndrome. We demonstrate that the accumulation of parasitised red blood cells (pRBCs) in brain capillaries is a specific feature of ECM that is not observed during mild murine malaria infections. Critically, we show that individual pRBCs appear to occlude murine brain capillaries during ECM. As pRBC-mediated congestion of brain microvessels is a hallmark of HCM, this suggests that the impact of parasite accumulation on cerebral blood flow may ultimately be similar in mice and humans during ECM and HCM, respectively. Additionally, we demonstrate that cerebrovascular CD8+ T-cells appear to co-localise with accumulated pRBCs, an event that corresponds with development of widespread vascular leakage. As in HCM, we show that vascular leakage is not dependent on extensive vascular destruction. Instead, we show that vascular leakage is associated with alterations in transcellular and paracellular transport mechanisms. Finally, as in HCM, we observed axonal injury and demyelination in ECM adjacent to diverse vasculopathies. Collectively, our data therefore shows that, despite very different presentation, and apparently distinct mechanisms, of parasite accumulation, there appear to be a number of comparable features of cerebral pathology in mice and in humans during ECM and HCM, respectively. Thus, when used appropriately, the ECM model may be useful for studying specific pathological features of HCM.


Asunto(s)
Encéfalo/patología , Encéfalo/parasitología , Modelos Animales de Enfermedad , Malaria Cerebral/patología , Malaria Cerebral/parasitología , Animales , Eritrocitos/parasitología , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Plasmodium berghei
10.
J Immunol ; 197(8): 3152-3164, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27630165

RESUMEN

CD4+ T cells that produce IFN-γ are the source of host-protective IL-10 during primary infection with a number of different pathogens, including Plasmodium spp. The fate of these CD4+IFN-γ+IL-10+ T cells following clearance of primary infection and their subsequent influence on the course of repeated infections is, however, presently unknown. In this study, utilizing IFN-γ-yellow fluorescent protein (YFP) and IL-10-GFP dual reporter mice, we show that primary malaria infection-induced CD4+YFP+GFP+ T cells have limited memory potential, do not stably express IL-10, and are disproportionately lost from the Ag-experienced CD4+ T cell memory population during the maintenance phase postinfection. CD4+YFP+GFP+ T cells generally exhibited a short-lived effector rather than effector memory T cell phenotype postinfection and expressed high levels of PD-1, Lag-3, and TIGIT, indicative of cellular exhaustion. Consistently, the surviving CD4+YFP+GFP+ T cell-derived cells were unresponsive and failed to proliferate during the early phase of secondary infection. In contrast, CD4+YFP+GFP- T cell-derived cells expanded rapidly and upregulated IL-10 expression during secondary infection. Correspondingly, CD4+ T cells were the major producers within an accelerated and amplified IL-10 response during the early stage of secondary malaria infection. Notably, IL-10 exerted quantitatively stronger regulatory effects on innate and CD4+ T cell responses during primary and secondary infections, respectively. The results in this study significantly improve our understanding of the durability of IL-10-producing CD4+ T cells postinfection and provide information on how IL-10 may contribute to optimized parasite control and prevention of immune-mediated pathology during repeated malaria infections.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Interleucina-10/metabolismo , Malaria/inmunología , Plasmodium/inmunología , Animales , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD4-Positivos/trasplante , Supervivencia Celular , Células Cultivadas , Humanos , Inmunosenescencia , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
11.
Infect Immun ; 84(1): 34-46, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26459508

RESUMEN

Immune-mediated pathology in interleukin-10 (IL-10)-deficient mice during blood-stage malaria infection typically manifests in nonlymphoid organs, such as the liver and lung. Thus, it is critical to define the cellular sources of IL-10 in these sensitive nonlymphoid compartments during infection. Moreover, it is important to determine if IL-10 production is controlled through conserved or disparate molecular programs in distinct anatomical locations during malaria infection, as this may enable spatiotemporal tuning of the regulatory immune response. In this study, using dual gamma interferon (IFN-γ)-yellow fluorescent protein (YFP) and IL-10-green fluorescent protein (GFP) reporter mice, we show that CD4(+) YFP(+) T cells are the major source of IL-10 in both lymphoid and nonlymphoid compartments throughout the course of blood-stage Plasmodium yoelii infection. Mature splenic CD4(+) YFP(+) GFP(+) T cells, which preferentially expressed high levels of CCR5, were capable of migrating to and seeding the nonlymphoid tissues, indicating that the systemically distributed host-protective cells have a common developmental history. Despite exhibiting comparable phenotypes, CD4(+) YFP(+) GFP(+) T cells from the liver and lung produced significantly larger quantities of IL-10 than their splenic counterparts, showing that the CD4(+) YFP(+) GFP(+) T cells exert graded functions in distinct tissue locations during infection. Unexpectedly, given the unique environmental conditions within discrete nonlymphoid and lymphoid organs, we show that IL-10 production by CD4(+) YFP(+) T cells is controlled systemically during malaria infection through IL-27 receptor signaling that is supported after CD4(+) T cell priming by ICOS signaling. The results in this study substantially improve our understanding of the systemic IL-10 response to malaria infection, particularly within sensitive nonlymphoid organs.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucina-10/inmunología , Interleucinas/inmunología , Malaria/inmunología , Traslado Adoptivo , Animales , Proteínas Bacterianas/genética , Linfocitos T CD4-Positivos/trasplante , Proteínas Fluorescentes Verdes/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-10/genética , Hígado/inmunología , Proteínas Luminiscentes/genética , Pulmón/inmunología , Malaria/parasitología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasmodium yoelii/inmunología , Bazo/inmunología
12.
PLoS Pathog ; 11(11): e1005210, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26562533

RESUMEN

There is significant evidence that brain-infiltrating CD8+ T cells play a central role in the development of experimental cerebral malaria (ECM) during Plasmodium berghei ANKA infection of C57BL/6 mice. However, the mechanisms through which they mediate their pathogenic activity during malaria infection remain poorly understood. Utilizing intravital two-photon microscopy combined with detailed ex vivo flow cytometric analysis, we show that brain-infiltrating T cells accumulate within the perivascular spaces of brains of mice infected with both ECM-inducing (P. berghei ANKA) and non-inducing (P. berghei NK65) infections. However, perivascular T cells displayed an arrested behavior specifically during P. berghei ANKA infection, despite the brain-accumulating CD8+ T cells exhibiting comparable activation phenotypes during both infections. We observed T cells forming long-term cognate interactions with CX3CR1-bearing antigen presenting cells within the brains during P. berghei ANKA infection, but abrogation of this interaction by targeted depletion of the APC cells failed to prevent ECM development. Pathogenic CD8+ T cells were found to colocalize with rare apoptotic cells expressing CD31, a marker of endothelial cells, within the brain during ECM. However, cellular apoptosis was a rare event and did not result in loss of cerebral vasculature or correspond with the extensive disruption to its integrity observed during ECM. In summary, our data show that the arrest of T cells in the perivascular compartments of the brain is a unique signature of ECM-inducing malaria infection and implies an important role for this event in the development of the ECM-syndrome.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Malaria Cerebral/inmunología , Malaria Falciparum/microbiología , Parasitemia/inmunología , Plasmodium berghei/inmunología , Animales , Linfocitos T CD8-positivos/parasitología , Modelos Animales de Enfermedad , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...