Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38826328

RESUMEN

Glycosylphosphatidylinositol (GPI) anchor protein modification in Plasmodium species is well known and represents the principal form of glycosylation in these organisms. The structure and biosynthesis of GPI anchors of Plasmodium spp. has been primarily studied in the asexual blood stage of P. falciparum and is known to contain the typical conserved GPI structure of EtN-P-Man3GlcN-PI. Here, we have investigated the circumsporozoite protein (CSP) for the presence of a GPI-anchor. CSP is the major surface protein of Plasmodium sporozoites, the infective stage of the malaria parasite. While it is widely assumed that CSP is a GPI-anchored cell surface protein, compelling biochemical evidence for this supposition is absent. Here, we employed metabolic labeling and mass-spectrometry based approaches to confirm the presence of a GPI anchor in CSP. Biosynthetic radiolabeling of CSP with [ 3 H]-palmitic acid and [ 3 H]-ethanolamine, with the former being base-labile and therefore ester-linked, provided strong evidence for the presence of a GPI anchor on CSP, but these data alone were not definitive. To provide further evidence, immunoprecipitated CSP was analyzed for presence of myo -inositol (a characteristic component of GPI anchor) using strong acid hydrolysis and GC-MS for a highly sensitive and quantitative detection. The single ion monitoring (SIM) method for GC-MS analysis confirmed the presence of the myo -inositol component in CSP. Taken together, these data provide confidence that the long-assumed presence of a GPI anchor on this important parasite protein is correct.

2.
Nat Commun ; 15(1): 748, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38272943

RESUMEN

Malaria is initiated when infected anopheline mosquitoes inoculate sporozoites as they probe for blood. It is thought that all infected mosquitoes are equivalent in terms of their infectious potential, with parasite burden having no role in transmission success. In this study, using mosquitoes harboring the entire range of salivary gland sporozoite loads observed in the field, we demonstrate a strong and highly significant correlation between mosquito parasite burden and inoculum size. We then link the inoculum data to oocyst counts, the most commonly-used metric to assess mosquito infection in the field, and determine the efficiency with which oocyst sporozoites enter mosquito salivary glands. Taken together our data support the conclusion that mosquitoes with higher parasite burdens are more likely to initiate infection and contribute to onward transmission. Overall these data may account for some of the unexplained heterogeneity in transmission and enable more precise benchmarks for transmission-blocking interventions.


Asunto(s)
Anopheles , Malaria , Parásitos , Plasmodium , Animales , Esporozoítos , Anopheles/parasitología , Malaria/parasitología
3.
Antimicrob Agents Chemother ; 66(9): e0041822, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-35943271

RESUMEN

As the malaria parasite becomes resistant to every drug that we develop, the identification and development of novel drug candidates are essential. Many studies have screened compounds designed to target the clinically important blood stages. However, if we are to shrink the malaria map, new drugs that block the transmission of the parasite are needed. Sporozoites are the infective stage of the malaria parasite, transmitted to the mammalian host as mosquitoes probe for blood. Sporozoite motility is critical to their ability to exit the inoculation site and establish infection, and drug-like compounds targeting motility are effective at blocking infection in the rodent malaria model. In this study, we established a moderate-throughput motility assay for sporozoites of the human malaria parasite Plasmodium falciparum, enabling us to screen the 400 drug-like compounds from the pathogen box provided by the Medicines for Malaria Venture for their activity. Compounds exhibiting inhibitory effects on P. falciparum sporozoite motility were further assessed for transmission-blocking activity and asexual-stage growth. Five compounds had a significant inhibitory effect on P. falciparum sporozoite motility in the nanomolar range. Using membrane feeding assays, we demonstrate that four of these compounds had inhibitory activity against the transmission of P. falciparum to the mosquito. Interestingly, of the four compounds with inhibitory activity against both transmission stages, three are known kinase inhibitors. Together with a previous study that found that several of these compounds could inhibit asexual blood-stage parasite growth, our findings provide new antimalarial drug candidates that have multistage activity.


Asunto(s)
Anopheles , Antimaláricos , Malaria Falciparum , Malaria , Animales , Anopheles/parasitología , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Humanos , Malaria/prevención & control , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Mamíferos , Plasmodium falciparum , Esporozoítos
4.
Nat Commun ; 12(1): 7046, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857782

RESUMEN

Reconstruction of heterogeneity through single cell transcriptional profiling has greatly advanced our understanding of the spatial liver transcriptome in recent years. However, global transcriptional differences across lobular units remain elusive in physical space. Here, we apply Spatial Transcriptomics to perform transcriptomic analysis across sectioned liver tissue. We confirm that the heterogeneity in this complex tissue is predominantly determined by lobular zonation. By introducing novel computational approaches, we enable transcriptional gradient measurements between tissue structures, including several lobules in a variety of orientations. Further, our data suggests the presence of previously transcriptionally uncharacterized structures within liver tissue, contributing to the overall spatial heterogeneity of the organ. This study demonstrates how comprehensive spatial transcriptomic technologies can be used to delineate extensive spatial gene expression patterns in the liver, indicating its future impact for studies of liver function, development and regeneration as well as its potential in pre-clinical and clinical pathology.


Asunto(s)
Heterogeneidad Genética , Hígado/metabolismo , Transcriptoma , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Eritroblastos/citología , Eritroblastos/metabolismo , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Hepatocitos/citología , Hepatocitos/metabolismo , Macrófagos del Hígado/citología , Macrófagos del Hígado/metabolismo , Hígado/citología , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Anotación de Secuencia Molecular , Neutrófilos/citología , Neutrófilos/metabolismo
5.
Sci Transl Med ; 13(599)2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162751

RESUMEN

Immunoglobulin (Ig)A antibodies play a critical role in protection against mucosal pathogens. However, the role of serum IgA in immunity to nonmucosal pathogens, such as Plasmodium falciparum, is poorly characterized, despite being the second most abundant isotype in blood after IgG. Here, we investigated the circulating IgA response in humans to P. falciparum sporozoites that are injected into the skin by mosquitoes and migrate to the liver via the bloodstream to initiate malaria infection. We found that circulating IgA was induced in three independent sporozoite-exposed cohorts: individuals living in an endemic region in Mali, malaria-naïve individuals immunized intravenously with three large doses of irradiated sporozoites, and malaria-naïve individuals exposed to a single controlled mosquito bite infection. Mechanistically, we found evidence in an animal model that IgA responses were induced by sporozoites at dermal inoculation sites. From malaria-resistant individuals, we isolated several IgA monoclonal antibodies that reduced liver parasite burden in mice. One antibody, MAD2-6, bound to a conserved epitope in the amino terminus of the P. falciparum circumsporozoite protein, the dominant protein on the sporozoite surface. Crystal structures of this antibody revealed a unique mode of binding whereby two Fabs simultaneously bound either side of the target peptide. This study reveals a role for circulating IgA in malaria and identifies the amino terminus of the circumsporozoite protein as a target of functional antibodies.


Asunto(s)
Anticuerpos Antiprotozoarios , Inmunoglobulina A , Malaria , Animales , Anticuerpos Antiprotozoarios/inmunología , Humanos , Inmunoglobulina A/inmunología , Malaria/inmunología , Ratones , Plasmodium falciparum , Proteínas Protozoarias , Esporozoítos
6.
J Leukoc Biol ; 110(4): 617-628, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34028876

RESUMEN

Protective cytotoxic and proinflammatory cytokine responses by NK cells impact the outcome of infections by Toxoplasma gondii, a common parasite in humans and other vertebrates. However, T. gondii can also sequester within NK cells and downmodulate their effector functions. Recently, the implication of GABA signaling in infection and inflammation-related responses of mononuclear phagocytes and T cells has become evident. Yet, the role of GABAergic signaling in NK cells has remained unknown. Here, we report that human and murine NK cells synthesize and secrete GABA in response to infection challenge. Parasitized NK cells secreted GABA, whereas activation stimuli, such as IL-12/IL-18 or parasite lysates, failed to induce GABA secretion. GABA secretion by NK cells was associated to a transcriptional up-regulation of GABA synthesis enzymes (glutamate decarboxylases [GAD65/67]) and was abrogated by GAD inhibition. Further, NK cells expressed GABA-A receptor subunits and GABA signaling regulators, with transcriptional modulations taking place upon challenge with T. gondii. Exogenous GABA and GABA-containing supernatants from parasitized dendritic cells (DCs) impacted NK cell function by reducing the degranulation and cytotoxicity of NK cells. Conversely, GABA-containing supernatants from NK cells enhanced the migratory responses of parasitized DCs. This enhanced DC migration was abolished by GABA-A receptor antagonism or GAD inhibition and was reconstituted by exogenous GABA. Jointly, the data show that NK cells are GABAergic cells and that GABA hampers NK cell cytotoxicity in vitro. We hypothesize that GABA secreted by parasitized immune cells modulates the immune responses to T. gondii infection.


Asunto(s)
Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/parasitología , Transducción de Señal , Toxoplasma/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Muerte Celular , Degranulación de la Célula/fisiología , Línea Celular , Movimiento Celular , Células Dendríticas/parasitología , Humanos , Células Asesinas Naturales/fisiología , Ratones Endogámicos C57BL , Transcripción Genética
7.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33653959

RESUMEN

Despite the critical role of Plasmodium sporozoites in malaria transmission, we still know little about the mechanisms underlying their development in mosquitoes. Here, we use single-cell RNA sequencing to characterize the gene expression profiles of 16,038 Plasmodium berghei sporozoites isolated throughout their development from midgut oocysts to salivary glands, and from forced salivation experiments. Our results reveal a succession of tightly regulated changes in gene expression occurring during the maturation of sporozoites and highlight candidate genes that could play important roles in oocyst egress, sporozoite motility, and the mechanisms underlying the invasion of mosquito salivary glands and mammalian hepatocytes. In addition, the single-cell data reveal extensive transcriptional heterogeneity among parasites isolated from the same anatomical site, suggesting that Plasmodium development in mosquitoes is asynchronous and regulated by intrinsic as well as environmental factors. Finally, our analyses show a decrease in transcriptional activity preceding the translational repression observed in mature sporozoites and associated with their quiescent state in salivary glands, followed by a rapid reactivation of the transcriptional machinery immediately upon salivation.


Asunto(s)
Anopheles/parasitología , Regulación de la Expresión Génica , Plasmodium berghei/metabolismo , Glándulas Salivales/parasitología , Esporozoítos/metabolismo , Transcripción Genética , Animales , Ratones
8.
EMBO Mol Med ; 13(4): e11796, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33750026

RESUMEN

Malaria infection starts with the injection of Plasmodium sporozoites into the host's skin. Sporozoites are motile and move in the skin to find and enter blood vessels to be carried to the liver. Here, we present the first characterization of P. falciparum sporozoites in vivo, analyzing their motility in mouse skin and human skin xenografts and comparing their motility to two rodent malaria species. These data suggest that in contrast to the liver and blood stages, the skin is not a species-specific barrier for Plasmodium. Indeed, P. falciparum sporozoites enter blood vessels in mouse skin at similar rates to the rodent malaria parasites. Furthermore, we demonstrate that antibodies targeting sporozoites significantly impact the motility of P. falciparum sporozoites in mouse skin. Though the sporozoite stage is a validated vaccine target, vaccine trials have been hampered by the lack of good animal models for human malaria parasites. Pre-clinical screening of next-generation vaccines would be significantly aided by the in vivo platform we describe here, expediting down-selection of candidates prior to human vaccine trials.


Asunto(s)
Vacunas contra la Malaria , Malaria Falciparum , Malaria , Animales , Humanos , Microscopía Intravital , Ratones , Plasmodium falciparum , Roedores , Esporozoítos
9.
Elife ; 92020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-33179597

RESUMEN

Gamma-aminobutyric acid (GABA) serves diverse biological functions in prokaryotes and eukaryotes, including neurotransmission in vertebrates. Yet, the role of GABA in the immune system has remained elusive. Here, a comprehensive characterization of human and murine myeloid mononuclear phagocytes revealed the presence of a conserved and tightly regulated GABAergic machinery with expression of GABA metabolic enzymes and transporters, GABA-A receptors and regulators, and voltage-dependent calcium channels. Infection challenge with the common coccidian parasites Toxoplasma gondii and Neospora caninum activated GABAergic signaling in phagocytes. Using gene silencing and pharmacological modulators in vitro and in vivo in mice, we identify the functional determinants of GABAergic signaling in parasitized phagocytes and demonstrate a link to calcium responses and migratory activation. The findings reveal a regulatory role for a GABAergic signaling machinery in the host-pathogen interplay between phagocytes and invasive coccidian parasites. The co-option of GABA underlies colonization of the host by a Trojan horse mechanism.


Asunto(s)
Fagocitos/metabolismo , Toxoplasma/fisiología , Toxoplasmosis Animal/parasitología , Ácido gamma-Aminobutírico/metabolismo , Traslado Adoptivo , Animales , Movimiento Celular , Células Cultivadas , Células Dendríticas/fisiología , Femenino , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR
10.
J Vis Exp ; (161)2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32716382

RESUMEN

Malaria remains one of the most important public health problems, causing significant morbidity and mortality. Malaria is a mosquito borne disease transmitted through an infectious bite from the female Anopheles mosquito. Malaria control will eventually rely on a multitude of approaches, which includes ways to block transmission to, through and from mosquitoes. To study mosquito stages of malaria parasites in the laboratory, we have optimized a protocol to culture highly infectious Plasmodium falciparum gametocytes, a parasite stage required for transmission from the human host to the mosquito vector. P. falciparum gametocytes mature through five morphologically distinct steps, which takes approximately 1-2 weeks. Gametocyte culture described in this protocol is completed in 15 days and are infectious to mosquitoes from days 15-18. These protocols were developed to maintain a continuous cycle of infection competent gametocytes and to maintain uninterrupted supply of mosquito stages of the parasite. Here, we describe the methodology of gametocyte culture and how to infect mosquitoes with these parasites using glass membrane feeders.


Asunto(s)
Anopheles/parasitología , Plasmodium falciparum , Animales , Femenino , Humanos , Malaria Falciparum , Membranas Artificiales , Mosquitos Vectores
11.
Sci Rep ; 9(1): 13131, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511546

RESUMEN

Malaria parasites have a complex life cycle that includes specialized stages for transmission between their mosquito and human hosts. These stages are an understudied part of the lifecycle yet targeting them is an essential component of the effort to shrink the malaria map. The human parasite Plasmodium falciparum is responsible for the majority of deaths due to malaria. Our goal was to generate transgenic P. falciparum lines that could complete the lifecycle and produce fluorescent transmission stages for more in-depth and high-throughput studies. Using zinc-finger nuclease technology to engineer an integration site, we generated three transgenic P. falciparum lines in which tdtomato or gfp were stably integrated into the genome. Expression was driven by either stage-specific peg4 and csp promoters or the constitutive ef1a promoter. Phenotypic characterization of these lines demonstrates that they complete the life cycle with high infection rates and give rise to fluorescent mosquito stages. The transmission stages are sufficiently bright for intra-vital imaging, flow cytometry and scalable screening of chemical inhibitors and inhibitory antibodies.


Asunto(s)
Proteínas Fluorescentes Verdes/genética , Proteínas Luminiscentes/genética , Malaria Falciparum/transmisión , Parásitos/genética , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Animales , Culicidae/parasitología , Citometría de Flujo/métodos , Ingeniería Genética/métodos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Estadios del Ciclo de Vida , Proteínas Luminiscentes/metabolismo , Malaria Falciparum/parasitología , Microscopía Fluorescente/métodos , Parásitos/crecimiento & desarrollo , Parásitos/fisiología , Fenotipo , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/fisiología , Proteínas Protozoarias/metabolismo , Proteína Fluorescente Roja
12.
Artículo en Inglés | MEDLINE | ID: mdl-30949457

RESUMEN

Toxoplasma gondii is a widespread obligate intracellular parasite that causes chronic infection and life-threatening acute infection in the central nervous system. Previous work identified Toxoplasma-infected microglia and astrocytes during reactivated infections in mice, indicating an implication of glial cells in acute toxoplasmic encephalitis. However, the mechanisms leading to the spread of Toxoplasma in the brain parenchyma remain unknown. Here, we report that, shortly after invasion by T. gondii tachyzoites, parasitized microglia, but not parasitized astrocytes, undergo rapid morphological changes and exhibit dramatically enhanced migration in 2-dimensional and 3-dimensional matrix confinements. Interestingly, primary microglia secreted the neurotransmitter γ-aminobutyric acid (GABA) in the supernatant as a consequence of T. gondii infection but not upon stimulation with LPS or heat-inactivated T. gondii. Further, microglia transcriptionally expressed components of the GABAergic machinery, including GABA-A receptor subunits, regulatory molecules and voltage-dependent calcium channels (VDCCs). Further, their transcriptional expression was modulated by challenge with T. gondii. Transcriptional analysis indicated that GABA was synthesized via both, the conventional pathway (glutamate decarboxylases GAD65 and GAD67) and a more recently characterized alternative pathway (aldehyde dehydrogenases ALDH2 and ALDH1a1). Pharmacological inhibitors targeting GABA synthesis, GABA-A receptors, GABA-A regulators and VDCC signaling inhibited Toxoplasma-induced hypermotility of microglia. Altogether, we show that primary microglia express a GABAergic machinery and that T. gondii induces hypermigration of microglia in a GABA-dependent fashion. We hypothesize that migratory activation of parasitized microglia by Toxoplasma may promote parasite dissemination in the brain parenchyma.


Asunto(s)
Movimiento Celular , Microglía/fisiología , Microglía/parasitología , Transducción de Señal , Toxoplasma/crecimiento & desarrollo , Toxoplasmosis/patología , Ácido gamma-Aminobutírico/metabolismo , Animales , Técnicas Citológicas , Perfilación de la Expresión Génica , Ratones Endogámicos C57BL , Modelos Teóricos
13.
PLoS Pathog ; 13(12): e1006739, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29216332

RESUMEN

The obligate intracellular parasite Toxoplasma gondii exploits cells of the immune system to disseminate. Upon T. gondii-infection, γ-aminobutyric acid (GABA)/GABAA receptor signaling triggers a hypermigratory phenotype in dendritic cells (DCs) by unknown signal transduction pathways. Here, we demonstrate that calcium (Ca2+) signaling in DCs is indispensable for T. gondii-induced DC hypermotility and transmigration in vitro. We report that activation of GABAA receptors by GABA induces transient Ca2+ entry in DCs. Murine bone marrow-derived DCs preferentially expressed the L-type voltage-dependent Ca2+ channel (VDCC) subtype Cav1.3. Silencing of Cav1.3 by short hairpin RNA or selective pharmacological antagonism of VDCCs abolished the Toxoplasma-induced hypermigratory phenotype. In a mouse model of toxoplasmosis, VDCC inhibition of adoptively transferred Toxoplasma-infected DCs delayed the appearance of cell-associated parasites in the blood circulation and reduced parasite dissemination to target organs. The present data establish that T. gondii-induced hypermigration of DCs requires signaling via VDCCs and that Ca2+ acts as a second messenger to GABAergic signaling via the VDCC Cav1.3. The findings define a novel motility-related signaling axis in DCs and unveil that interneurons and DCs share common GABAergic motogenic pathways. T. gondii employs GABAergic non-canonical pathways to induce host cell migration and facilitate dissemination.


Asunto(s)
Canales de Calcio Tipo L/inmunología , Señalización del Calcio , Células Dendríticas/inmunología , Receptores de GABA-A/inmunología , Toxoplasma/inmunología , Toxoplasmosis/inmunología , Traslado Adoptivo , Animales , Movimiento Celular , Células Cultivadas , Células Dendríticas/parasitología , GABAérgicos/inmunología , Ratones , Ratones Endogámicos C57BL , Toxoplasma/fisiología , Toxoplasmosis/parasitología , Ácido gamma-Aminobutírico/inmunología
14.
Infect Immun ; 85(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28760930

RESUMEN

The obligate intracellular parasite Toxoplasma gondii can actively infect any nucleated cell type, including cells from the immune system. The rapid transfer of T. gondii from infected dendritic cells to effector natural killer (NK) cells may contribute to the parasite's sequestration and shielding from immune recognition shortly after infection. However, subversion of NK cell functions, such as cytotoxicity or production of proinflammatory cytokines, such as gamma interferon (IFN-γ), upon parasite infection might also be beneficial to the parasite. In the present study, we investigated the effects of T. gondii infection on NK cells. In vitro, infected NK cells were found to be poor at killing target cells and had reduced levels of IFN-γ production. This could be attributed in part to the inability of infected cells to form conjugates with their target cells. However, even upon NK1.1 cross-linking of NK cells, the infected NK cells also exhibited poor degranulation and IFN-γ production. Similarly, NK cells infected in vivo were also poor at killing target cells and producing IFN-γ. Increased levels of transforming growth factor ß production, as well as increased levels of expression of SHP-1 in the cytosol of infected NK cells upon infection, were observed in infected NK cells. However, the phosphorylation of STAT4 was not altered in infected NK cells, suggesting that transcriptional regulation mediates the reduced IFN-γ production, which was confirmed by quantitative PCR. These data suggest that infection of NK cells by T. gondii impairs NK cell recognition of target cells and cytokine release, two mechanisms that independently could enhance T. gondii survival.


Asunto(s)
Inmunomodulación , Células Asesinas Naturales/microbiología , Células Asesinas Naturales/fisiología , Toxoplasma/inmunología , Toxoplasmosis Animal/inmunología , Animales , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Interacciones Huésped-Parásitos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Células Asesinas Naturales/inmunología , Ratones , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Factor de Transcripción STAT4/metabolismo , Toxoplasma/fisiología , Factor de Crecimiento Transformador beta/biosíntesis
15.
Nat Immunol ; 17(11): 1282-1290, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27618552

RESUMEN

Glioma cells recruit and exploit microglia (the resident immune cells of the brain) for their proliferation and invasion ability. The underlying molecular mechanism used by glioma cells to transform microglia into a tumor-supporting phenotype has remained elusive. We found that glioma-induced microglia conversion was coupled to a reduction in the basal activity of microglial caspase-3 and increased S-nitrosylation of mitochondria-associated caspase-3 through inhibition of thioredoxin-2 activity, and that inhibition of caspase-3 regulated microglial tumor-supporting function. Furthermore, we identified the activity of nitric oxide synthase 2 (NOS2, also known as iNOS) originating from the glioma cells as a driving stimulus in the control of microglial caspase-3 activity. Repression of glioma NOS2 expression in vivo led to a reduction in both microglia recruitment and tumor expansion, whereas depletion of microglial caspase-3 gene promoted tumor growth. Our results provide evidence that inhibition of the denitrosylation of S-nitrosylated procaspase-3 mediated by the redox protein Trx2 is a part of the microglial pro-tumoral activation pathway initiated by glioma cancer cells.


Asunto(s)
Caspasa 3/metabolismo , Glioma/metabolismo , Glioma/patología , Microglía/metabolismo , Fenotipo , Animales , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Activación Enzimática , Técnicas de Silenciamiento del Gen , Glioma/inmunología , Xenoinjertos , Humanos , Masculino , Ratones , Microglía/inmunología , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Tiorredoxinas/metabolismo , Carga Tumoral
16.
Cell Microbiol ; 18(11): 1537-1550, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27018989

RESUMEN

The obligate intracellular parasite Toxoplasma gondii exploits cells of the immune system to disseminate. Upon infection, parasitized dendritic cells (DCs) and microglia exhibit a hypermigratory phenotype in vitro that has been associated with enhancing parasite dissemination in vivo in mice. One unresolved question is how parasites commandeer parasitized cells to achieve systemic dissemination by a 'Trojan-horse' mechanism. By chromatography and mass spectrometry analyses, we identified an orthologue of the 14-3-3 protein family, T. gondii 14-3-3 (Tg14-3-3), as mediator of DC hypermotility. We demonstrate that parasite-derived polypeptide fractions enriched for Tg14-3-3 or recombinant Tg14-3-3 are sufficient to induce the hypermotile phenotype when introduced by protein transfection into murine DCs, human DCs or microglia. Further, gene transfer of Tg14-3-3 by lentiviral transduction induced hypermotility in primary human DCs. In parasites expressing Tg14-3-3 in a ligand-regulatable fashion, overexpression of Tg14-3-3 was correlated with induction of hypermotility in parasitized DCs. Localization studies in infected DCs identified Tg14-3-3 within the parasitophorous vacuolar space and a rapid recruitment of host cell 14-3-3 to the parasitophorous vacuole membrane. The present work identifies a determinant role for Tg14-3-3 in the induction of the migratory activation of immune cells by T. gondii. Collectively, the findings reveal Tg14-3-3 as a novel target for an intracellular pathogen that acts by hijacking the host cell's migratory properties to disseminate.


Asunto(s)
Proteínas 14-3-3/fisiología , Células Dendríticas/fisiología , Proteínas Protozoarias/fisiología , Toxoplasma/fisiología , Animales , Movimiento Celular , Células Cultivadas , Células Dendríticas/parasitología , Interacciones Huésped-Parásitos , Humanos , Ratones Endogámicos C57BL , Vacuolas/metabolismo , Vacuolas/parasitología
17.
PLoS One ; 10(9): e0139104, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26406763

RESUMEN

Toxoplasma gondii, an obligate intracellular parasite of humans and other warm-blooded vertebrates, invades a variety of cell types in the organism, including immune cells. Notably, dendritic cells (DCs) infected by T. gondii acquire a hypermigratory phenotype that potentiates parasite dissemination by a 'Trojan horse' type of mechanism in mice. Previous studies have demonstrated that, shortly after parasite invasion, infected DCs exhibit hypermotility in 2-dimensional confinements in vitro and enhanced transmigration in transwell systems. However, interstitial migration in vivo involves interactions with the extracellular matrix in a 3-dimensional (3D) space. We have developed a collagen matrix-based assay in a 96-well plate format that allows quantitative locomotion analyses of infected DCs in a 3D confinement over time. We report that active invasion of DCs by T. gondii tachyzoites induces enhanced migration of infected DCs in the collagen matrix. Parasites of genotype II induced superior DC migratory distances than type I parasites. Moreover, Toxoplasma-induced hypermigration of DCs was further potentiated in the presence of the CCR7 chemotactic cue CCL19. Blocking antibodies to integrins (CD11a, CD11b, CD18, CD29, CD49b) insignificantly affected migration of infected DCs in the 3D matrix, contrasting with their inhibitory effects on adhesion in 2D assays. Morphological analyses of infected DCs in the matrix were consistent with the acquisition of an amoeboid-like migratory phenotype. Altogether, the present data show that the Toxoplasma-induced hypermigratory phenotype in a 3D matrix is consistent with integrin-independent amoeboid DC migration with maintained responsiveness to chemotactic and chemokinetic cues. The data support the hypothesis that induction of amoeboid hypermigration and chemotaxis/chemokinesis in infected DCs potentiates the dissemination of T. gondii.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Colágeno/farmacología , Células Dendríticas/parasitología , Toxoplasma/fisiología , Toxoplasmosis/patología , Animales , Anticuerpos Bloqueadores/farmacología , Efecto Espectador/efectos de los fármacos , Bovinos , Adhesión Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Quimiocina CCL19/farmacología , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Humanos , Integrinas/metabolismo , Ratones , Fenotipo
18.
Cell Microbiol ; 15(10): 1735-52, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23534541

RESUMEN

Host cell manipulation is an important feature of the obligate intracellular parasite Toxoplasma gondii. Recent reports have shown that the tachyzoite stages subvert dendritic cells (DC) as a conduit for dissemination (Trojan horse) during acute infection. To examine the cellular basis of these processes, we performed a detailed analysis of the early events following tachyzoite invasion of human monocyte-derived DC. We demonstrate that within minutes after tachyzoite penetration, profound morphological changes take place in DC that coincide with a migratory activation. Active parasite invasion of DC led to cytoskeletal actin redistribution with loss of adhesive podosome structures and redistribution of integrins (CD18 and CD11c), that concurred with the onset of DC hypermotility in vitro. Inhibition of parasite rhoptry secretion and invasion, but not inhibition of parasite or host cell protein synthesis, abrogated the onset of morphological changes and hypermotility in DC dose-dependently. Also, infected DC, but not by-stander DC, exhibited upregulation of C-C chemokine receptor 7 (CCR7). Yet, the onset of parasite-induced DC hypermotility preceded chemotactic migratory responsesin vitro. Collectively, present data reveal that invasion of DC by T. gondii initiates a series of regulated events, including rapid cytoskeleton rearrangements, hypermotility and chemotaxis, that promote the migratory activation of DC.


Asunto(s)
Movimiento Celular , Citoesqueleto/metabolismo , Células Dendríticas/fisiología , Células Dendríticas/parasitología , Endocitosis , Interacciones Huésped-Patógeno , Toxoplasma/fisiología , Células Cultivadas , Quimiotaxis , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...