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












Base de datos
Intervalo de año de publicación
1.
Nature ; 623(7986): 397-405, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37914940

RESUMEN

Microglia are specialized brain-resident macrophages that arise from primitive macrophages colonizing the embryonic brain1. Microglia contribute to multiple aspects of brain development, but their precise roles in the early human brain remain poorly understood owing to limited access to relevant tissues2-6. The generation of brain organoids from human induced pluripotent stem cells recapitulates some key features of human embryonic brain development7-10. However, current approaches do not incorporate microglia or address their role in organoid maturation11-21. Here we generated microglia-sufficient brain organoids by coculturing brain organoids with primitive-like macrophages generated from the same human induced pluripotent stem cells (iMac)22. In organoid cocultures, iMac differentiated into cells with microglia-like phenotypes and functions (iMicro) and modulated neuronal progenitor cell (NPC) differentiation, limiting NPC proliferation and promoting axonogenesis. Mechanistically, iMicro contained high levels of PLIN2+ lipid droplets that exported cholesterol and its esters, which were taken up by NPCs in the organoids. We also detected PLIN2+ lipid droplet-loaded microglia in mouse and human embryonic brains. Overall, our approach substantially advances current human brain organoid approaches by incorporating microglial cells, as illustrated by the discovery of a key pathway of lipid-mediated crosstalk between microglia and NPCs that leads to improved neurogenesis.


Asunto(s)
Encéfalo , Colesterol , Células Madre Pluripotentes Inducidas , Microglía , Células-Madre Neurales , Neurogénesis , Organoides , Animales , Humanos , Ratones , Encéfalo/citología , Encéfalo/metabolismo , Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Microglía/citología , Microglía/metabolismo , Organoides/citología , Organoides/metabolismo , Colesterol/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Axones , Proliferación Celular , Ésteres/metabolismo , Gotas Lipídicas/metabolismo
3.
FASEB J ; 35(12): e21997, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34719814

RESUMEN

The deadliest complication of infection by Plasmodium parasites, cerebral malaria, accounts for the majority of malarial fatalities. Although our understanding of the cellular and molecular mechanisms underlying the pathology remains incomplete, recent studies support the contribution of systemic and neuroinflammation as the cause of cerebral edema and blood-brain barrier (BBB) dysfunction. All Plasmodium species encode an orthologue of the innate cytokine, Macrophage Migration Inhibitory Factor (MIF), which functions in mammalian biology to regulate innate responses. Plasmodium MIF (PMIF) similarly signals through the host MIF receptor CD74, leading to an enhanced inflammatory response. We investigated the PMIF-CD74 interaction in the onset of experimental cerebral malaria (ECM) and liver stage Plasmodium development by using a combination of CD74 deficient (Cd74-/- ) hosts and PMIF deficient parasites. Cd74-/- mice were found to be protected from ECM and the protection was associated with the inability of brain microvessels to present parasite antigen to sequestered and pathogenic Plasmodium-specific CD8+ T cells. Infection of WT hosts with PMIF-deficient sporozoites or infection of Cd74-/- hosts with WT sporozoites impacted the survival of infected hepatocytes and subsequently reduced blood-stage associated inflammation, contributing to protection from ECM. We recapitulated these finding with a novel pharmacologic PMIF-selective antagonist that reduced PMIF/CD74 signaling and fully protected mice from ECM. These findings reveal a conserved mechanism for Plasmodium usurpation of host CD74 signaling and suggest a tractable approach for new pharmacologic intervention.


Asunto(s)
Antígenos de Diferenciación de Linfocitos B/química , Linfocitos T CD8-positivos/inmunología , Antígenos de Histocompatibilidad Clase II/química , Inflamación/prevención & control , Hígado/patología , Factores Inhibidores de la Migración de Macrófagos/antagonistas & inhibidores , Malaria Cerebral/prevención & control , Plasmodium berghei/fisiología , Animales , Antígenos de Diferenciación de Linfocitos B/fisiología , Antígenos de Histocompatibilidad Clase II/fisiología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Hígado/inmunología , Hígado/parasitología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Malaria Cerebral/etiología , Malaria Cerebral/metabolismo , Malaria Cerebral/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
4.
Immunity ; 54(9): 2101-2116.e6, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34469775

RESUMEN

Tissue macrophages are immune cells whose phenotypes and functions are dictated by origin and niches. However, tissues are complex environments, and macrophage heterogeneity within the same organ has been overlooked so far. Here, we used high-dimensional approaches to characterize macrophage populations in the murine liver. We identified two distinct populations among embryonically derived Kupffer cells (KCs) sharing a core signature while differentially expressing numerous genes and proteins: a major CD206loESAM- population (KC1) and a minor CD206hiESAM+ population (KC2). KC2 expressed genes involved in metabolic processes, including fatty acid metabolism both in steady-state and in diet-induced obesity and hepatic steatosis. Functional characterization by depletion of KC2 or targeted silencing of the fatty acid transporter Cd36 highlighted a crucial contribution of KC2 in the liver oxidative stress associated with obesity. In summary, our study reveals that KCs are more heterogeneous than anticipated, notably describing a subpopulation wired with metabolic functions.


Asunto(s)
Antígenos CD36/metabolismo , Macrófagos del Hígado/metabolismo , Hígado/metabolismo , Obesidad/metabolismo , Estrés Oxidativo/fisiología , Animales , Ratones
5.
iScience ; 24(5): 102482, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34113823

RESUMEN

Clinically important broadly reactive B cells evolve during multiple infections, with B cells re-activated after secondary infection differing from B cells activated after a primary infection. Here we studied CD27highCD38high plasmablasts from patients with a primary or secondary dengue virus infection. Three transcriptionally and functionally distinct clusters were identified. The largest cluster 0/1 was plasma cell-related, with cells coding for serotype cross-reactive antibodies of the IgG1 isotype, consistent with memory B cell activation during an extrafollicular response. Cells in clusters 2 and 3 expressed low levels of antibody genes and high levels of genes associated with oxidative phosphorylation, EIF2 pathway, and mitochondrial dysfunction. Clusters 2 and 3 showed a transcriptional footprint of T cell help, in line with activation from naive B cells or memory B cells. Our results contribute to the understanding of the parallel B cell activation events that occur in humans after natural primary and secondary infection.

6.
Elife ; 92020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32066522

RESUMEN

In malaria, rosetting is described as a phenomenon where an infected erythrocyte (IRBC) is attached to uninfected erythrocytes (URBC). In some studies, rosetting has been associated with malaria pathogenesis. Here, we have identified a new type of rosetting. Using a step-by-step approach, we identified IGFBP7, a protein secreted by monocytes in response to parasite stimulation, as a rosette-stimulator for Plasmodium falciparum- and P. vivax-IRBC. IGFBP7-mediated rosette-stimulation was rapid yet reversible. Unlike type I rosetting that involves direct interaction of rosetting ligands on IRBC and receptors on URBC, the IGFBP7-mediated, type II rosetting requires two additional serum factors, namely von Willebrand factor and thrombospondin-1. These two factors interact with IGFBP7 to mediate rosette formation by the IRBC. Importantly, the IGFBP7-induced type II rosetting hampers phagocytosis of IRBC by host phagocytes.


Malaria is a life-threatening disease transmitted by mosquitoes infected with Plasmodium parasites. Part of the parasite life cycle happens inside human red blood cells. The surface of an infected red blood cell is coated with parasite proteins, which attract the attention of white blood cells called monocytes. These immune cells circulate in the bloodstream and use a process called phagocytosis to essentially 'eat' any infected cells they encounter. However, the monocytes cannot always reach the infected cells. Some of the proteins made by the parasites make the infected red blood cells stickier than normal. This allows the infected red blood cells to surround themselves in a protective cage of uninfected red blood cells. Known as "rosettes" because of their flower-like shape, these cages seem to protect the infected cells from attack by the immune system. Lee et al. noticed that adding white blood cells to parasite-infected red blood cells made them clump together more, but it was unclear exactly how and why this happened. To find out, Lee et al. took fluid from around monocytes grown in the laboratory and added it to red blood cells infected with Plasmodium parasites. This made the cells clump together, suggesting that something in the fluid may potentially be alerting the parasites to impending immune attack. The fluid contained almost 700 different molecules, and Lee et al. narrowed down their investigations to the five most likely candidates. Interfering with the activities of these five proteins revealed that one ­ a protein IGFBP7 ­ not only alerted the parasites but also helped them to form the rosettes. It turns out that the parasites appear to use IGFBP7 like a bridge, linking it to two other human proteins to stick red blood cells together. Once the rosettes had formed, the monocytes were unable to eat the infected blood cells by themselves. Instead several monocytes had to work together as a team to consume the whole rosette. Further research is now needed to shed light on this interaction between malaria parasites and human cells. Such research would be particularly relevant in the clinical setting, since some previous studies has linked the forming of rosettes to the severity of disease for malaria.


Asunto(s)
Eritrocitos/parasitología , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Fagocitosis , Plasmodium falciparum/fisiología , Plasmodium vivax/fisiología , Medios de Cultivo , Humanos , Ligandos , Pruebas de Neutralización , Células THP-1
7.
Front Immunol ; 9: 1875, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30154793

RESUMEN

Cerebral malaria is a complex neurological syndrome caused by an infection with Plasmodium falciparum parasites and is exclusively attributed to a series of host-parasite interactions at the pathological blood-stage of infection. In contrast, the preceding intra-hepatic phase of replication is generally considered clinically silent and thereby excluded from playing any role in the development of neurological symptoms. In this study, however, we present an antigen PbmaLS_05 that is presented to the host immune system by both pre-erythrocytic and intra-erythrocytic stages and contributes to the development of cerebral malaria in mice. Although deletion of the endogenous PbmaLS_05 prevented the development of experimental cerebral malaria (ECM) in susceptible mice after both sporozoite and infected red blood cell (iRBC) infections, we observed significant differences in contribution of the host immune response between both modes of inoculation. Moreover, PbmaLS_05-specific CD8+ T cells contributed to the development of ECM after sporozoite but not iRBC-infection, suggesting that pre-erythrocytic antigens like PbmaLS_05 can also contribute to the development of cerebral symptoms. Our data thus highlight the importance of the natural route of infection in the study of ECM, with potential implications for vaccine and therapeutic strategies against malaria.


Asunto(s)
Antígenos de Protozoos/inmunología , Susceptibilidad a Enfermedades , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Plasmodium berghei/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Reactividad Cruzada/inmunología , Modelos Animales de Enfermedad , Expresión Génica , Genes Protozoarios , Genes Reporteros , Estadios del Ciclo de Vida , Imagen por Resonancia Magnética , Malaria Cerebral/diagnóstico , Malaria Cerebral/patología , Ratones , Plasmodium berghei/genética , Plasmodium berghei/crecimiento & desarrollo
8.
PLoS One ; 13(2): e0192717, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29438386

RESUMEN

Malaria ranks among the most important infectious diseases worldwide and affects mostly people living in tropical countries. Mechanisms involved in disease progression are still not fully understood and specific treatments that might interfere with cerebral malaria (CM) are limited. Here we show that administration of doxycycline (DOX) prevented experimental CM (ECM) in Plasmodium berghei ANKA (PbA)-infected C57BL/6 wildtype (WT) mice in an IL-10-independent manner. DOX-treated mice showed an intact blood-brain barrier (BBB) and attenuated brain inflammation. Importantly, if WT mice were infected with a 20-fold increased parasite load, they could be still protected from ECM if they received DOX from day 4-6 post infection, despite similar parasitemia compared to control-infected mice that did not receive DOX and developed ECM. Infiltration of T cells and cytotoxic responses were reduced in brains of DOX-treated mice. Analysis of brain tissue by RNA-array revealed reduced expression of chemokines and tumour necrosis factor (TNF) in brains of DOX-treated mice. Furthermore, DOX-administration resulted in brains of the mice in reduced expression of matrix metalloproteinase 2 (MMP2) and granzyme B, which are both factors associated with ECM pathology. Systemic interferon gamma production was reduced and activated peripheral T cells accumulated in the spleen in DOX-treated mice. Our results suggest that DOX targeted inflammatory processes in the central nervous system (CNS) and prevented ECM by impaired brain access of effector T cells in addition to its anti-parasitic effect, thereby expanding the understanding of molecular events that underlie DOX-mediated therapeutic interventions.


Asunto(s)
Antimaláricos/farmacología , Doxiciclina/farmacología , Malaria Cerebral/prevención & control , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/inmunología , Quimiocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inflamación/inmunología , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Interleucina-10/metabolismo , Activación de Linfocitos/efectos de los fármacos , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Parasitemia/inmunología , Parasitemia/prevención & control , Plasmodium berghei/inmunología , Plasmodium berghei/patogenicidad , Bazo/efectos de los fármacos , Bazo/inmunología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología
9.
Cell Microbiol ; 20(5): e12819, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29281764

RESUMEN

Host immune response has a key role in controlling the progression of malaria infection. In the well-established murine model of experimental cerebral malaria (ECM) with Plasmodium berghei ANKA infection, proinflammatory Th1 and CD8+ T cell response are essential for disease development. Interferon regulatory factor 1 (IRF1) is a transcription factor that promotes Th1 responses, and its absence was previously shown to protect from ECM death. Yet the exact mechanism of protection remains unknown. Here we demonstrated that IRF1-deficient mice (IRF1 knockout) were protected from ECM death despite displaying early neurological signs. Resistance to ECM death was a result of reduced parasite sequestration and pathogenic CD8+ T cells in the brain. Further analysis revealed that IRF1 deficiency suppress interferon-γ production and delayed CD8+ T cell proliferation. CXCR3 expression was found to be decreased in pathogenic CD8+ T cells, which limited their migration to the brain. In addition, reduced expression of adhesion molecules by brain endothelial cells hampered leucocyte retention in the brain. Taken together, these factors limited sequestration of pathogenic CD8+ T cells and consequently its ability to induce extensive damage to the blood-brain barrier.


Asunto(s)
Factor 1 Regulador del Interferón/genética , Malaria Cerebral/genética , Plasmodium berghei/patogenicidad , Receptores CXCR3/genética , Animales , Encéfalo/microbiología , Encéfalo/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Movimiento Celular/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/microbiología , Ratones , Ratones Noqueados
10.
EMBO Mol Med ; 10(1): 121-138, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29113976

RESUMEN

Arboviral diseases have risen significantly over the last 40 years, increasing the risk of co-infection with other endemic disease such as malaria. However, nothing is known about the impact arboviruses have on the host response toward heterologous pathogens during co-infection. Here, we investigate the effects of Chikungunya virus (CHIKV) co-infection on the susceptibility and severity of malaria infection. Using the Plasmodium berghei ANKA (PbA) experimental cerebral malaria (ECM) model, we show that concurrent co-infection induced the most prominent changes in ECM manifestation. Concurrent co-infection protected mice from ECM mortality without affecting parasite development in the blood. This protection was mediated by the alteration of parasite-specific CD8+ T-cell trafficking through an IFNγ-mediated mechanism. Co-infection with CHIKV induced higher splenic IFNγ levels that lead to high local levels of CXCL9 and CXCL10. This induced retention of CXCR3-expressing pathogenic CD8+ T cells in the spleen and prevented their migration to the brain. This then averts all downstream pathogenic events such as parasite sequestration in the brain and disruption of blood-brain barrier that prevents ECM-induced mortality in co-infected mice.


Asunto(s)
Encéfalo/patología , Linfocitos T CD8-positivos/patología , Fiebre Chikungunya/patología , Virus Chikungunya/fisiología , Coinfección/patología , Malaria Cerebral/patología , Plasmodium berghei/fisiología , Animales , Encéfalo/parasitología , Encéfalo/virología , Linfocitos T CD8-positivos/parasitología , Linfocitos T CD8-positivos/virología , Movimiento Celular , Fiebre Chikungunya/parasitología , Fiebre Chikungunya/virología , Coinfección/parasitología , Coinfección/virología , Femenino , Malaria Cerebral/parasitología , Malaria Cerebral/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropatología , Factores Protectores
11.
JCI Insight ; 2(8)2017 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-28422757

RESUMEN

Zika virus (ZIKV) infections have been linked with neurological complications and congenital Zika syndrome. Given the high level of homology between ZIKV and the related flavivirus dengue virus (DENV), we investigated the level of cross-reactivity with ZIKV using a panel of DENV human mAbs. A majority of the mAbs showed binding to ZIKV virions, with several exhibiting neutralizing capacities against ZIKV in vitro. Three of the best ZIKV-neutralizing mAbs were found to recognize diverse epitopes on the envelope (E) glycoprotein: the highly conserved fusion-loop peptide, a conformation-specific epitope on the E monomer, and a quaternary epitope on the virion surface. The most potent ZIKV-neutralizing mAb (SIgN-3C) was assessed in 2 type I interferon receptor-deficient (IFNAR-/-) mouse models of ZIKV infection. Treatment of adult nonpregnant mice with SIgN-3C rescued mice from virus-induced weight loss and mortality. The SIgN-3C variant with Leu-to-Ala mutations in the Fc region (SIgN-3C-LALA) did not induce antibody-dependent enhancement (ADE) in vitro but provided similar levels of protection in vivo. In pregnant ZIKV-infected IFNAR-/- mice, treatment with SIgN-3C or SIgN-3C-LALA significantly reduced viral load in the fetal organs and placenta and abrogated virus-induced fetal growth retardation. Therefore, SIgN-3C-LALA holds promise as a ZIKV prophylactic and therapeutic agent.

12.
Nat Protoc ; 10(12): 2016-26, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26562622

RESUMEN

We have recently demonstrated that brain endothelial cells cross-present parasite antigen during mouse experimental cerebral malaria (ECM). Here we describe a 2-d protocol to detect cross-presentation by isolating the brain microvessels and incubating them with a reporter cell line that expresses lacZ upon detection of the relevant peptide-major histocompatibility complex. After X-gal staining, a typical positive result consists of hundreds of blue spots, compared with fewer than 20 spots from a naive brain. The assay is generalizable to other disease contexts by using reporter cells that express appropriate specific T cell receptors. Also described is the protocol for culturing endothelial cells from brain microvessels isolated from naive mice. After 7-10 d, an in vitro cross-presentation assay can be performed by adding interferon-γ, antigen (e.g., Plasmodium berghei-infected red blood cells) and reporter cells in sequence over 3 d. This is useful for comparing different antigen forms or for probing the effects of various interventions.


Asunto(s)
Presentación de Antígeno , Encéfalo/inmunología , Células Endoteliales/inmunología , Microvasos/inmunología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/citología , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/citología , Complejo Mayor de Histocompatibilidad , Malaria/inmunología , Ratones , Ratones Endogámicos C57BL , Microvasos/citología , Plasmodium berghei/inmunología , Receptores de Antígenos de Linfocitos T/inmunología
13.
PLoS Pathog ; 11(6): e1004963, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26046849

RESUMEN

In the murine model of cerebral malaria caused by P. berghei ANKA (PbA), parasite-specific CD8+ T cells directly induce pathology and have long been hypothesized to kill brain endothelial cells that have internalized PbA antigen. We previously reported that brain microvessel fragments from infected mice cross-present PbA epitopes, using reporter cells transduced with epitope-specific T cell receptors. Here, we confirm that endothelial cells are the population responsible for cross-presentation in vivo, not pericytes or microglia. PbA antigen cross-presentation by primary brain endothelial cells in vitro confers susceptibility to killing by CD8+ T cells from infected mice. IFNγ stimulation is required for brain endothelial cross-presentation in vivo and in vitro, which occurs by a proteasome- and TAP-dependent mechanism. Parasite strains that do not induce cerebral malaria were phagocytosed and cross-presented less efficiently than PbA in vitro. The main source of antigen appears to be free merozoites, which were avidly phagocytosed. A human brain endothelial cell line also phagocytosed P. falciparum merozoites. Besides being the first demonstration of cross-presentation by brain endothelial cells, our results suggest that interfering with merozoite phagocytosis or antigen processing may be effective strategies for cerebral malaria intervention.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Protozoos/inmunología , Encéfalo/inmunología , Reactividad Cruzada/inmunología , Células Endoteliales/inmunología , Malaria Cerebral/inmunología , Animales , Encéfalo/parasitología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos C57BL
14.
Infect Immun ; 82(11): 4854-64, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25156726

RESUMEN

CD8(+) T cells play a pathogenic role in the development of murine experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA) infection in C57BL/6 mice. Only a limited number of CD8(+) epitopes have been described. Here, we report the identification of a new epitope from the bergheilysin protein recognized by PbA-specific CD8(+) T cells. Induction and functionality of these specific CD8(+) T cells were investigated in parallel with previously reported epitopes, using new tools such as tetramers and reporter cell lines that were developed for this study. We demonstrate that CD8(+) T cells of diverse specificities induced during PbA infection share many characteristics. They express cytolytic markers (gamma interferon [IFN-γ], granzyme B) and chemokine receptors (CXCR3, CCR5) and damage the blood-brain barrier in vivo. Our earlier finding that brain microvessels in mice infected with PbA, but not with non-ECM-causing strains, cross-presented a shared epitope was generalizable to these additional epitopes. Suppressing the induction of specific CD8(+) T cells through tolerization with a high-dose peptide injection was unable to confer protection against ECM, suggesting that CD8(+) T cells of other specificities participate in this process. The tools that we developed can be used to further investigate the heterogeneity of CD8(+) T cell responses that are involved in ECM.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Malaria Cerebral/complicaciones , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Barrera Hematoencefálica , Epítopos , Regulación de la Expresión Génica/inmunología , Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Ratones , Ratones Endogámicos C57BL , Plasmodium berghei/inmunología
15.
EMBO Mol Med ; 5(7): 984-99, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23681698

RESUMEN

Cerebral malaria is a devastating complication of Plasmodium falciparum infection. Its pathogenesis is complex, involving both parasite- and immune-mediated events. CD8(+) T cells play an effector role in murine experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA) infection. We have identified a highly immunogenic CD8 epitope in glideosome-associated protein 50 that is conserved across rodent malaria species. Epitope-specific CD8(+) T cells are induced during PbA infection, migrating to the brain just before neurological signs manifest. They are functional, cytotoxic and can damage the blood-brain barrier in vivo. Such CD8(+) T cells are also found in the brain during infection with parasite strains/species that do not induce neuropathology. We demonstrate here that PbA infection causes brain microvessels to cross-present parasite antigen, while non-ECM-causing parasites do not. Further, treatment with fast-acting anti-malarial drugs before the onset of ECM reduces parasite load and thus antigen presentation in the brain, preventing ECM death. Thus our data suggest that combined therapies targeting both the parasite and host antigen-presenting cells may improve the outcome of CM patients.


Asunto(s)
Antígenos de Protozoos/inmunología , Encéfalo/irrigación sanguínea , Encéfalo/parasitología , Linfocitos T CD8-positivos/inmunología , Reactividad Cruzada , Malaria Cerebral/inmunología , Plasmodium berghei/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Protozoos/análisis , Antimaláricos/uso terapéutico , Barrera Hematoencefálica/inmunología , Barrera Hematoencefálica/parasitología , Barrera Hematoencefálica/patología , Encéfalo/inmunología , Encéfalo/patología , Linfocitos T CD8-positivos/química , Linfocitos T CD8-positivos/parasitología , Reactividad Cruzada/efectos de los fármacos , Femenino , Humanos , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/parasitología , Malaria Cerebral/patología , Ratones , Ratones Endogámicos C57BL , Microvasos/inmunología , Microvasos/parasitología , Microvasos/patología , Datos de Secuencia Molecular , Carga de Parásitos , Plasmodium berghei/efectos de los fármacos , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología
16.
Anal Biochem ; 416(1): 141-3, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21530478

RESUMEN

Directional cloning of complementary DNA (cDNA) primed by oligo(dT) is commonly achieved by appending a restriction site to the primer, whereas the second strand is synthesized through the combined action of RNase H and Escherichia coli DNA polymerase I (PolI). Although random primers provide more uniform and complete coverage, directional cloning with the same strategy is highly inefficient. We report that phosphorothioate linkages protect the tail sequence appended to random primers from the 5'→3' exonuclease activity of PolI. We present a simple strategy for constructing a random-primed cDNA library using the efficient, size-independent, and seamless In-Fusion cloning method instead of restriction enzymes.


Asunto(s)
Cartilla de ADN/química , Cartilla de ADN/genética , ADN Complementario/genética , Biblioteca de Genes , Fosfatos/química , Enzimas de Restricción del ADN
17.
J Mol Biol ; 401(1): 84-96, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20540948

RESUMEN

The rugged protein sequence-function landscape complicates efforts, both in nature and in the laboratory, to evolve protein function. Protein library diversification must strike a balance between sufficient variegation to thoroughly sample alternative functionality versus the probability of mutant destabilization below an expressible threshold. In this work, we explore the sequence-function landscape in the context of screening for molecular recognition from an Ig scaffold library. The fibronectin type III domain is used to explore the impact of two sequence diversification strategies: (a) partial wild-type conservation at structurally important positions within the paratope region and (b) tailored amino acid composition mimicking antibody binding-site composition at putative paratope positions. Structurally important positions within the paratope region were identified through stability, structural, and phylogenetic analyses and partially or fully conserved in sequence. To achieve tailored antibody-like diversity, we designed a set of skewed nucleotide mixtures yielding codons approximately matching the distribution observed in antibody complementarity-determining regions without incurring the expense of triphosphoramidite-based construction. These design elements were explored via comparison of three library designs: a random library, a library with wild-type bias in the DE loop only and tyrosine-serine diversity elsewhere, and a library with wild-type bias at 11 positions and the antibody-inspired amino acid distribution. Using pooled libraries for direct competition in a single tube, selection and maturation of binders to seven targets yielded 19 of 21 clones that originated from the structurally biased, tailored-diversity library design. Sequence analysis of the selected clones supports the importance of both tailored compositional diversity and structural bias. In addition, selection of both well and poorly expressed clones from two libraries further elucidated the impact of structural bias.


Asunto(s)
Anticuerpos/química , Anticuerpos/metabolismo , Regiones Determinantes de Complementariedad/química , Regiones Determinantes de Complementariedad/metabolismo , Secuencia de Aminoácidos , Animales , Afinidad de Anticuerpos , Unión Competitiva , Células Cultivadas , Humanos , Modelos Moleculares , Biblioteca de Péptidos , Filogenia , Unión Proteica/fisiología , Ingeniería de Proteínas/métodos , Estabilidad Proteica , Saccharomyces cerevisiae , Análisis de Secuencia de Proteína/métodos , Relación Estructura-Actividad
18.
J Immunother ; 31(7): 607-19, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18600183

RESUMEN

Saccharomyces cerevisiae stimulates dendritic cells (DCs) and represents a promising candidate for cancer vaccine development. Effective cross-presentation of antigen delivered to DCs is necessary for successful induction of cellular immunity. Here, we present a yeast-based vaccine approach that is independent of yeast's ability to express the chosen antigen, which is instead produced separately and conjugated to the yeast cell wall. The conjugation method is site-specific (based on the SNAP-tag) and designed to facilitate antigen release in the DC phagosome and subsequent translocation for cross-presentation. We demonstrate that nonsite-specific chemical conjugation of the same protein hinders cross-presentation. Phagosomal antigen release was further expedited through the insertion of the invariant chain ectodomain as a linker, which is rapidly cleaved by Cathepsin S. The dose of delivered antigen was increased in several ways: by using yeast strains with higher surface amine densities, by using yeast hulls (cell wall fragments) instead of whole cells, and by conjugating multiple layers of antigen. The novel multilayer conjugation scheme takes advantage of Sfp phosphopantetheinyl transferase and remains site-specific; it enables the antigen dose to grow linearly with the number of layers. We show that whole yeast cells coated with 1 layer of the cancer-testis antigen NY-ESO-1 and yeast hulls bearing 3 layers were able to cross-prime naive CD8 T cells in vitro, with the latter resulting in higher frequencies of antigen-specific cells after 10 days. This cross-presentation-efficient antigen conjugation scheme is not limited to yeast and can readily be applied toward the development of other particulate vaccines.


Asunto(s)
Presentación de Antígeno/inmunología , Vacunas contra el Cáncer/inmunología , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Escherichia coli , Saccharomyces cerevisiae , Aminación , Antígenos de Diferenciación de Linfocitos B/inmunología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Vesículas Cubiertas/inmunología , Citomegalovirus/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/microbiología , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos/inmunología , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , O(6)-Metilguanina-ADN Metiltransferasa/química , O(6)-Metilguanina-ADN Metiltransferasa/inmunología , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Fosfoproteínas/genética , Fosfoproteínas/inmunología , Transporte de Proteínas/inmunología , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Saccharomyces cerevisiae/inmunología , Neoplasias Testiculares/inmunología , Neoplasias Testiculares/terapia , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología
19.
J Immunol ; 180(3): 1576-83, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18209053

RESUMEN

Cross-presentation of exogenous Ags in MHC class I molecules by dendritic cells is the underlying basis for many developing immunotherapies and vaccines. In the phagosome-to-cytosol pathway, Ags in phagocytosed particles must become freely soluble before being exported to the cytosol, but the kinetics of this process has yet to be fully appreciated. We demonstrate with a yeast vaccine model that the rate of Ag release in the phagosome directly affects cross-presentation efficiency, with an apparent time limit of approximately 25 min postphagocytosis for Ag release to be productive. Ag expressed on the yeast surface is cross-presented much more efficiently than Ag trapped in the yeast cytosol by the cell wall. The cross-presentation efficiency of yeast surface-displayed Ag can be increased by the insertion of linkers susceptible to cleavage in the early phagosome. Ags indirectly attached to yeast through Ab fragments are less efficiently cross-presented when the Ab dissociation rate is extremely slow.


Asunto(s)
Antígenos/inmunología , Reactividad Cruzada , Antígenos de Histocompatibilidad Clase I/inmunología , Fagosomas/inmunología , Secuencia de Aminoácidos , Antígenos/análisis , Antígenos/genética , Antígenos de Histocompatibilidad Clase I/análisis , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Región Variable de Inmunoglobulina , Cinética , Datos de Secuencia Molecular , Levaduras/genética , Levaduras/inmunología
20.
Protein Expr Purif ; 48(2): 232-42, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16563796

RESUMEN

NY-ESO-1 is a highly immunogenic tumor antigen and a promising vaccine candidate in cancer immunotherapy. Access to purified protein both for vaccine formulations and for monitoring antigen-specific immune responses is vital to vaccine development. Currently available recombinant Escherichia coli-derived NY-ESO-1 is isolated from inclusion bodies as a complex protein mixture and efforts to improve the purity of this antigen are required, especially for later-stage clinical trials. Using yeast cell surface display and fluorescence activated cell sorting techniques, we have engineered an NY-ESO-1 variant (NY-ESO-L5; C(75)A C(76)A C(78)A L(153)H) with a 100x improved display level on yeast compared to the wild-type protein. This mutant can be effectively produced as an Aga2p-fusion and purified in soluble form directly from the yeast cell wall. In the process, we have identified the epitope recognized by anti-NY-ESO-1 mAb E978 (79-87, GARGPESRL). The availability of an alternative expression host for this important antigen will help avoid artifactual false positive tests of patient immune response due to reaction against expression-host-specific contaminants.


Asunto(s)
Antígenos de Neoplasias/genética , Evolución Molecular Dirigida/métodos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Antígenos de Neoplasias/aislamiento & purificación , Escherichia coli , Técnicas Genéticas , Humanos , Masculino , Proteínas de la Membrana/aislamiento & purificación , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...