Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Immunol ; 195(8): 3890-900, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26371254

RESUMEN

Cytokines are key regulators of adequate immune responses to infection with Mycobacterium tuberculosis. We demonstrate that the p110δ catalytic subunit of PI3K acts as a downstream effector of the TLR family member RP105 (CD180) in promoting mycobacteria-induced cytokine production by macrophages. Our data show that the significantly reduced release of TNF and IL-6 by RP105(-/-) macrophages during mycobacterial infection was not accompanied by diminished mRNA or protein expression. Mycobacteria induced comparable activation of NF-κB and p38 MAPK signaling in wild-type (WT) and RP105(-/-) macrophages. In contrast, mycobacteria-induced phosphorylation of Akt was abrogated in RP105(-/-) macrophages. The p110δ-specific inhibitor, Cal-101, and small interfering RNA-mediated knockdown of p110δ diminished mycobacteria-induced TNF secretion by WT but not RP105(-/-) macrophages. Such interference with p110δ activity led to reduced surface-expressed TNF in WT but not RP105(-/-) macrophages, while leaving TNF mRNA and protein expression unaffected. Activity of Bruton's tyrosine kinase was required for RP105-mediated activation of Akt phosphorylation and TNF release by mycobacteria-infected macrophages. These data unveil a novel innate immune signaling axis that orchestrates key cytokine responses of macrophages and provide molecular insight into the functions of RP105 as an innate immune receptor for mycobacteria.


Asunto(s)
Antígenos CD/inmunología , Fosfatidilinositol 3-Quinasa Clase I/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Antígenos CD/genética , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/genética , Inhibidores Enzimáticos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/inmunología , Purinas/farmacología , Quinazolinonas/farmacología , Tuberculosis/genética , Tuberculosis/patología , Factor de Necrosis Tumoral alfa/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología
2.
Cell Microbiol ; 17(6): 767-84, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25488819

RESUMEN

Upon infection, Legionella pneumophila uses the Dot/Icm type IV secretion system to translocate effector proteins from the Legionella-containing vacuole (LCV) into the host cell cytoplasm. The effectors target a wide array of host cellular processes that aid LCV biogenesis, including the manipulation of membrane trafficking. In this study, we used a hidden Markov model screen to identify two novel, non-eukaryotic soluble NSF attachment protein receptor (SNARE) homologs: the bacterial Legionella SNARE effector A (LseA) and viral SNARE homolog A proteins. We characterized LseA as a Dot/Icm effector of L. pneumophila, which has close homology to the Qc-SNARE subfamily. The lseA gene was present in multiple sequenced L. pneumophila strains including Corby and was well distributed among L. pneumophila clinical and environmental isolates. Employing a variety of biochemical, cell biological and microbiological techniques, we found that farnesylated LseA localized to membranes associated with the Golgi complex in mammalian cells and LseA interacted with a subset of Qa-, Qb- and R-SNAREs in host cells. Our results suggested that LseA acts as a SNARE protein and has the potential to regulate or mediate membrane fusion events in Golgi-associated pathways.


Asunto(s)
Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno , Legionella pneumophila/fisiología , Imitación Molecular , Proteínas Solubles de Unión al Factor Sensible a la N-Etilmaleimida/metabolismo , Factores de Virulencia/metabolismo , Animales , Línea Celular , Células Epiteliales/microbiología , Humanos , Macrófagos/microbiología , Ratones , Homología de Secuencia de Aminoácido
3.
J Immunol ; 192(8): 3709-18, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24634490

RESUMEN

Organ-specific immunity is a feature of many infectious diseases, including visceral leishmaniasis caused by Leishmania donovani. Experimental visceral leishmaniasis in genetically susceptible mice is characterized by an acute, resolving infection in the liver and chronic infection in the spleen. CD4+ T cell responses are critical for the establishment and maintenance of hepatic immunity in this disease model, but their role in chronically infected spleens remains unclear. In this study, we show that dendritic cells are critical for CD4+ T cell activation and expansion in all tissue sites examined. We found that FTY720-mediated blockade of T cell trafficking early in infection prevented Ag-specific CD4+ T cells from appearing in lymph nodes, but not the spleen and liver, suggesting that early CD4+ T cell priming does not occur in liver-draining lymph nodes. Extended treatment with FTY720 over the first month of infection increased parasite burdens, although this associated with blockade of lymphocyte egress from secondary lymphoid tissue, as well as with more generalized splenic lymphopenia. Importantly, we demonstrate that CD4+ T cells are required for the establishment and maintenance of antiparasitic immunity in the liver, as well as for immune surveillance and suppression of parasite outgrowth in chronically infected spleens. Finally, although early CD4+ T cell priming appeared to occur most effectively in the spleen, we unexpectedly revealed that protective CD4+ T cell-mediated hepatic immunity could be generated in the complete absence of all secondary lymphoid tissues.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Leishmania donovani/inmunología , Leishmaniasis Visceral/inmunología , Animales , Antígenos de Protozoos/inmunología , Linfocitos T CD4-Positivos/efectos de los fármacos , Células Dendríticas/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Clorhidrato de Fingolimod , Inmunosupresores/farmacología , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/parasitología , Activación de Linfocitos/inmunología , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/inmunología , Tejido Linfoide/parasitología , Ratones , Ratones Noqueados , Glicoles de Propileno/farmacología , Esfingosina/análogos & derivados , Esfingosina/farmacología , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/parasitología
4.
Immunol Cell Biol ; 92(3): 275-86, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24343664

RESUMEN

Rho GTPases are required for many cellular events such as adhesion, motility, and membrane trafficking. Here we show that in macrophages, the Rho GTPases Rac1 and Cdc42 are involved in lamellipodia and filopodia formation, respectively, and that both of these Rho GTPases are essential for the efficient surface delivery of tumor necrosis factor (TNF) to the plasma membrane following TLR4 stimulation. We have previously demonstrated intracellular trafficking of TNF via recycling endosomes in lipopolysaccharide (LPS)-activated macrophages. Here, we further define a specific role for Rac1 in intracellular TNF trafficking, demonstrating impairment in TNF release following TLR4 stimulation in the presence of a Rac inhibitor, in cells expressing a dominant negative (DN) form of Rac1, and following small interfering RNA (siRNA) knockdown of Rac1. Rac1 activity was required for TNF trafficking but not for TLR4 signaling following LPS stimulation. Reduced TNF secretion was due to a defect in Rac1 activity, but not of the closely related Rho GTPase Rac2, demonstrated by the additional use of macrophages derived from Rac2-deficient mice. Labeling recycling endosomes by the uptake of fluorescent transferrin enabled us to show that Rac1 was required for the final stages of TNF trafficking and delivery from recycling endosomes to the plasma membrane. Thus, actin remodeling by the Rho GTPase Rac1 is required for TNF cell surface delivery and release from macrophages.


Asunto(s)
Endocitosis , Endosomas/metabolismo , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Transporte de Proteínas/efectos de los fármacos , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Pironas/farmacología , Quinolinas/farmacología , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína RCA2 de Unión a GTP
5.
PLoS Pathog ; 7(10): e1002279, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21998581

RESUMEN

LIGHT (TNFSF14) is a member of the TNF superfamily involved in inflammation and defence against infection. LIGHT signals via two cell-bound receptors; herpes virus entry mediator (HVEM) and lymphotoxin-beta receptor (LTßR). We found that LIGHT is critical for control of hepatic parasite growth in mice with visceral leishmaniasis (VL) caused by infection with the protozoan parasite Leishmania donovani. LIGHT-HVEM signalling is essential for early dendritic cell IL-12/IL-23p40 production, and the generation of IFNγ- and TNF-producing T cells that control hepatic infection. However, we also discovered that LIGHT-LTßR interactions suppress anti-parasitic immunity in the liver in the first 7 days of infection by mechanisms that restrict both CD4(+) T cell function and TNF-dependent microbicidal mechanisms. Thus, we have identified distinct roles for LIGHT in infection, and show that manipulation of interactions between LIGHT and its receptors may be used for therapeutic advantage.


Asunto(s)
Inmunidad Celular , Leishmania donovani/patogenicidad , Leishmaniasis Visceral/patología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Proliferación Celular/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-12/biosíntesis , Interleucina-23/biosíntesis , Leishmania donovani/inmunología , Leishmaniasis Visceral/inmunología , Leishmaniasis Visceral/parasitología , Hígado/parasitología , Hígado/patología , Receptor beta de Linfotoxina/inmunología , Receptor beta de Linfotoxina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal , Linfocitos T/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
6.
J Immunother Cancer ; 11(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36634920

RESUMEN

BACKGROUND: Pixatimod is a unique activator of the Toll-like Receptor 9 pathway. This phase I trial evaluated safety, efficacy and pharmacodynamics of pixatimod and PD-1 inhibitor nivolumab in immunologically cold cancers. METHODS: 3+3 dose escalation with microsatellite stable metastatic colorectal cancer (MSS mCRC) and metastatic pancreatic ductal adenocarcinoma (mPDAC) expansion cohorts. Participants received pixatimod once weekly as a 1-hour intravenous infusion plus nivolumab every 2 weeks. Objectives included assessment of safety, antitumor activity, pharmacodynamics, and pharmacokinetic profile. RESULTS: Fifty-eight participants started treatment. The maximum tolerated dose of pixatimod was 25 mg in combination with 240 mg nivolumab, which was used in the expansion phases of the study. Twenty-one grade 3-5 treatment-related adverse events were reported in 12 participants (21%); one participant receiving 50 mg pixatimod/nivolumab had a treatment-related grade 5 AE. The grade 3/4 rate in the MSS mCRC cohort (n=33) was 12%. There were no responders in the mPDAC cohort (n=18). In the MSS mCRC cohort, 25 participants were evaluable (initial postbaseline assessment scans >6 weeks); of these, three participants had confirmed partial responses (PR) and eight had stable disease (SD) for at least 9 weeks. Clinical benefit (PR+SD) was associated with lower Pan-Immune-Inflammation Value and plasma IL-6 but increased IP-10 and IP-10/IL-8 ratio. In an MSS mCRC participant with PR as best response, increased infiltration of T cells, dendritic cells, and to a lesser extent NK cells, were evident 5 weeks post-treatment. CONCLUSIONS: Pixatimod is well tolerated at 25 mg in combination with nivolumab. The efficacy signal and pharmacodynamic changes in MSS mCRC warrants further investigation. TRIAL REGISTRATION NUMBER: NCT05061017.


Asunto(s)
Adenocarcinoma , Neoplasias Colorrectales , Humanos , Nivolumab/farmacología , Nivolumab/uso terapéutico , Receptor Toll-Like 9 , Quimiocina CXCL10 , Adenocarcinoma/patología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Inhibidores de la Angiogénesis/uso terapéutico , Repeticiones de Microsatélite , Neoplasias Pancreáticas
7.
J Immunol ; 184(5): 2583-92, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20139272

RESUMEN

Chronic infectious diseases and cancers are often associated with suboptimal effector T cell responses. Enhancement of T cell costimulatory signals has been extensively studied for cancer immunotherapy but not so for the treatment of infectious disease. The few previous attempts at this strategy using infection models have lacked cellular specificity, with major immunoregulatory mechanisms or innate immune cells also being targeted. In this study, we examined the potential of promoting T cell responses via the glucocorticoid-induced TNF receptor (GITR) family-related protein in a murine model of visceral leishmaniasis. GITR stimulation during established infection markedly improved antiparasitic immunity. This required CD4(+) T cells, TNF, and IFN-gamma, but crucially, was independent of regulatory T (Treg) cells. GITR stimulation enhanced CD4(+) T cell expansion without modulating Treg cell function or protecting conventional CD4(+) T cells from Treg cell suppression. GITR stimulation substantially improved the efficacy of a first-line visceral leishmaniasis drug against both acute hepatic infection and chronic infection in the spleen, demonstrating its potential to improve clinical outcomes. This study identifies a novel strategy to therapeutically enhance CD4(+) T cell-mediated antiparasitic immunity and, importantly, achieves this goal without impairment of Treg cell function.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Leishmaniasis Visceral/inmunología , Receptores de Factor de Crecimiento Nervioso/inmunología , Receptores del Factor de Necrosis Tumoral/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Gluconato de Sodio Antimonio/administración & dosificación , Antiprotozoarios/administración & dosificación , Apoptosis/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Sinergismo Farmacológico , Femenino , Proteína Relacionada con TNFR Inducida por Glucocorticoide , Inmunidad Celular/inmunología , Interferón gamma/metabolismo , Leishmania donovani/efectos de los fármacos , Leishmania donovani/crecimiento & desarrollo , Leishmania donovani/inmunología , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/parasitología , Hígado/inmunología , Hígado/metabolismo , Hígado/parasitología , Parasitosis Hepáticas/inmunología , Parasitosis Hepáticas/metabolismo , Parasitosis Hepáticas/parasitología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Factor de Crecimiento Nervioso/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
J Immunol ; 184(9): 4852-62, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20368273

RESUMEN

NK cells are renowned for their ability to kill virally infected or transformed host cells by release of cytotoxic granules containing granzymes and perforin. NK cells also have important regulatory capabilities chiefly mediated by secretion of cytokines, such as IFN-gamma and TNF. The secretory pathway for the release of cytokines in NK cells is unknown. In this study, we show localization and trafficking of IFN-gamma and TNF in human NK cells in compartments and vesicles that do not overlap with perforin or other late endosome granule markers. Cytokines in post-Golgi compartments colocalized with markers of the recycling endosome (RE). REs are functionally required for cytokine release because inactivation of REs or mutation of RE-associated proteins Rab11 and vesicle-associated membrane protein-3 blocked cytokine surface delivery and release. In contrast, REs are not needed for release of perforin from preformed granules but may be involved at earlier stages of granule maturation. These findings suggest a new role for REs in orchestrating secretion in NK cells. We show that the cytokines IFN-gamma and TNF are trafficked and secreted via a different pathway than perforin. Although perforin granules are released in a polarized fashion at lytic synapses, distinct carriers transport both IFN-gamma and TNF to points all over the cell surface, including within the synapse, for nonpolarized release.


Asunto(s)
Gránulos Citoplasmáticos/inmunología , Gránulos Citoplasmáticos/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Perforina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto , Compartimento Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Polaridad Celular/inmunología , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad/métodos , Endosomas/inmunología , Endosomas/metabolismo , Humanos , Sinapsis Inmunológicas/metabolismo , Interferón gamma/biosíntesis , Células K562 , Activación de Linfocitos/inmunología , Perforina/biosíntesis , Transporte de Proteínas/inmunología , Vesículas Secretoras/inmunología , Vesículas Secretoras/metabolismo , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
9.
J Immunol ; 185(6): 3632-42, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20720206

RESUMEN

Cerebral malaria is a severe complication of malaria. Sequestration of parasitized RBCs in brain microvasculature is associated with disease pathogenesis, but our understanding of this process is incomplete. In this study, we examined parasite tissue sequestration in an experimental model of cerebral malaria (ECM). We show that a rapid increase in parasite biomass is strongly associated with the induction of ECM, mediated by IFN-gamma and lymphotoxin alpha, whereas TNF and IL-10 limit this process. Crucially, we discovered that host CD4(+) and CD8(+) T cells promote parasite accumulation in vital organs, including the brain. Modulation of CD4(+) T cell responses by helminth coinfection amplified CD4(+) T cell-mediated parasite sequestration, whereas vaccination could generate CD4(+) T cells that reduced parasite biomass and prevented ECM. These findings provide novel insights into immune-mediated mechanisms of ECM pathogenesis and highlight the potential of T cells to both prevent and promote infectious diseases.


Asunto(s)
Malaria Cerebral/inmunología , Malaria Cerebral/parasitología , Plasmodium berghei/inmunología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Encéfalo/parasitología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD4-Positivos/patología , Modelos Animales de Enfermedad , Eritrocitos/inmunología , Eritrocitos/parasitología , Eritrocitos/patología , Femenino , Tracto Gastrointestinal/irrigación sanguínea , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/parasitología , Riñón/irrigación sanguínea , Riñón/inmunología , Riñón/parasitología , Hígado/irrigación sanguínea , Hígado/inmunología , Hígado/parasitología , Pulmón/irrigación sanguínea , Pulmón/inmunología , Pulmón/parasitología , Malaria Cerebral/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Especificidad de Órganos/inmunología , Plasmodium berghei/crecimiento & desarrollo , Índice de Severidad de la Enfermedad , Bazo/irrigación sanguínea , Bazo/inmunología , Bazo/parasitología
10.
Exp Cell Res ; 317(13): 1817-29, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21586284

RESUMEN

The ability of cells to adhere, spread and migrate is essential to many physiological processes, particularly in the immune system where cells must traffic to sites of inflammation and injury. By altering the levels of individual components of the VAMP3/Stx4/SNAP23 complex we show here that this SNARE complex regulates efficient macrophage adhesion, spreading and migration on fibronectin. During cell spreading this complex mediates the polarised exocytosis of VAMP3-positive recycling endosome membrane into areas of membrane expansion, where VAMP3's surface partner Q-SNARE complex Stx4/SNAP23 was found to accumulate. Lowering the levels of VAMP3 in spreading cells resulted in a more rounded cell morphology and most cells were found to be devoid of the typical ring-like podosome superstructures seen normally in spreading cells. In migrating cells lowering VAMP3 levels disrupted the polarised localisation of podosome clusters. The reduced trafficking of recycling endosome membrane to sites of cell spreading and the disorganised podosome localisation in migrating macrophages greatly reduced their ability to persistently migrate on fibronectin. Thus, this important SNARE complex facilitates macrophage adhesion, spreading, and persistent macrophage migration on fibronectin through the delivery of VAMP3-positive membrane with its cargo to expand the plasma membrane and to participate in organising adhesive podosome structures.


Asunto(s)
Movimiento Celular , Forma de la Célula , Macrófagos/citología , Macrófagos/metabolismo , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Proteína 3 de Membrana Asociada a Vesículas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Adhesión Celular , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL
11.
Physiology (Bethesda) ; 25(4): 218-29, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20699468

RESUMEN

Cytokine secretion is a widely studied process, although little is known regarding the specific mechanisms that regulate cytokine release. Recent findings have shed light on some of the precise molecular pathways that regulate the packaging of newly synthesized cytokines from immune cells. These findings begin to elucidate pathways and mechanisms that underpin cytokine release in all cells. In this article, we review the highlights of some of these novel discoveries.


Asunto(s)
Citocinas/metabolismo , Redes y Vías Metabólicas/fisiología , Vasos Sanguíneos/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Exocitosis/fisiología , Humanos , Proteínas SNARE/fisiología
12.
J Infect Dis ; 202(1): 117-24, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20500087

RESUMEN

BACKGROUND: Age and host genetics are important determinants of malaria severity. Lymphotoxin-alpha (LTalpha) has been associated with the development of cerebral malaria (CM) and other severe malaria (SM) syndromes. Mutations in genes regulating LTalpha production contribute to other acute vascular diseases and may contribute to malaria pathogenesis. METHODS: We tested the association between rs7291467, a single-nucleotide polymorphism (SNP) in the LTalpha-related gene encoding galectin-2 (LGALS2), disease severity, and function in a case-control study of ethnic Highland Papuan adults and children with SM (n = 380) and asymptomatic malaria-exposed controls (n = 356) originating from a non-malaria-endemic region but residing in a lowland malaria-endemic area of Papua, Indonesia. RESULTS: The LGALS2 SNP showed a significant association with susceptibility to SM (including CM), in children (odds ratio, 2.02 [95% confidence interval, 1.14-3.57]) but not in adults. In SM, the C allele at rs7291467 was associated with enhanced galectin-2 transcript levels. In a separate group of Tanzanian children originating from a malaria-endemic region, we found preservation of the major ancestral LGALS2 allele and no association with susceptibility to CM. CONCLUSIONS: Results suggest differences in the inflammatory contribution to the development of SM between children and adults in the same population and potential differences between individuals originating from malaria-endemic and non-malaria-endemic areas.


Asunto(s)
Galectina 2/genética , Malaria Falciparum/genética , Adolescente , Adulto , Distribución por Edad , Anciano , Estudios de Casos y Controles , Niño , Preescolar , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Indonesia/epidemiología , Lactante , Intrones , Malaria Falciparum/epidemiología , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Adulto Joven
13.
PLoS Pathog ; 4(9): e1000158, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18802456

RESUMEN

Vascular cell adhesion molecule-1 (VCAM-1) interacts with its major ligand very late antigen-4 (VLA-4) to mediate cell adhesion and transendothelial migration of leukocytes. We report an important role for VCAM-1/VLA-4 interactions in the generation of immune responses during experimental visceral leishmaniasis caused by Leishmania donovani. Our studies demonstrate that these molecules play no direct role in the recruitment of leukocytes to the infected liver, but instead contribute to IL-12p40-production by splenic CD8(+) dendritic cells (DC). Blockade of VCAM-1/VLA-4 interactions using whole antibody or anti-VCAM-1 Fab' fragments reduced IL-12p40 mRNA accumulation by splenic DC 5 hours after L. donovani infection. This was associated with reduced anti-parasitic CD4(+) T cell activation in the spleen and lowered hepatic IFNgamma, TNF and nitric oxide production by 14 days post infection. Importantly, these effects were associated with enhanced parasite growth in the liver in studies with either anti-VCAM-1 or anti-VLA-4 antibodies. These data indicate a role for VCAM-1 and VLA-4 in DC activation during infectious disease.


Asunto(s)
Células Dendríticas/metabolismo , Integrina alfa4beta1/inmunología , Subunidad p40 de la Interleucina-12/biosíntesis , Leishmaniasis Visceral/inmunología , Molécula 1 de Adhesión Celular Vascular/inmunología , Animales , Anticuerpos Antiprotozoarios/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Leishmania donovani , Hígado/parasitología , Activación de Linfocitos , Ratones , Ratones Endogámicos , Bazo/inmunología
14.
PLoS Pathog ; 4(2): e1000028, 2008 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-18463695

RESUMEN

We report that natural killer T (NKT) cells play only a minor physiological role in protection from Leishmania donovani infection in C57BL/6 mice. Furthermore, attempts at therapeutic activation of invariant NKT (iNKT) cells with alpha-galactosylceramide (alpha-GalCer) during L. donovani infection exacerbated, rather than ameliorated, experimental visceral leishmaniasis. The inability of alpha-GalCer to promote anti-parasitic immunity did not result from inefficient antigen presentation caused by infection because alpha-GalCer-loaded bone marrow-derived dendritic cells were also unable to improve disease resolution. The immune-dampening affect of alpha-GalCer correlated with a bias towards increased IL-4 production by iNKT cells following alpha-GalCer stimulation in infected mice compared to naïve controls. However, studies in IL-4-deficient mice, and IL-4 neutralisation in cytokine-sufficient mice revealed that alpha-GalCer-induced IL-4 production during infection had only a minor role in impaired parasite control. Analysis of liver cell composition following alpha-GalCer stimulation during an established L. donovani infection revealed important differences, predominantly a decrease in IFNgamma+ CD8+ T cells, compared with control-treated mice. Our data clearly illustrate the double-edged sword of NKT cell-based therapy, showing that in some circumstances, such as when sub-clinical or chronic infections exist, iNKT cell activation can have adverse outcomes.


Asunto(s)
Células Asesinas Naturales/inmunología , Leishmania donovani/patogenicidad , Leishmaniasis Visceral/inmunología , Subgrupos Linfocitarios/inmunología , Animales , Anticuerpos Bloqueadores/farmacología , Biomarcadores , Linfocitos T CD8-positivos , Modelos Animales de Enfermedad , Femenino , Galactosilceramidas/uso terapéutico , Silenciador del Gen , Interacciones Huésped-Parásitos , Factores Inmunológicos/uso terapéutico , Interferón gamma , Interleucina-4/deficiencia , Interleucina-4/inmunología , Interleucina-4/metabolismo , Leishmania donovani/inmunología , Leishmaniasis Visceral/tratamiento farmacológico , Leishmaniasis Visceral/patología , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/parasitología , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo
15.
Malar J ; 9: 302, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-21029472

RESUMEN

BACKGROUND: Severe malaria (SM) syndromes caused by Plasmodium falciparum infection result in major morbidity and mortality each year. However, only a fraction of P. falciparum infections develop into SM, implicating host genetic factors as important determinants of disease outcome. Previous studies indicate that tumour necrosis factor (TNF) and lymphotoxin alpha (LTα) may be important for the development of cerebral malaria (CM) and other SM syndromes. METHODS: An extensive analysis was conducted of single nucleotide polymorphisms (SNPs) in the TNF, LTA and LTB genes in highland Papuan children and adults, a population historically unexposed to malaria that has migrated to a malaria endemic region. Generated P-values for SNPs spanning the LTA/TNF/LTB locus were corrected for multiple testing of all the SNPs and haplotype blocks within the region tested through 10,000 permutations. A global P-value of < 0.05 was considered statistically significant. RESULTS: No associations between SNPs in the TNF/LTA/LTB locus and susceptibility to SM in highland Papuan children and adults were found. CONCLUSIONS: These results support the notion that unique selective pressure on the TNF/LTA/LTB locus in different populations has influenced the contribution of the gene products from this region to SM susceptibility.


Asunto(s)
Predisposición Genética a la Enfermedad , Linfotoxina-alfa/genética , Linfotoxina beta/genética , Malaria Falciparum/genética , Malaria Falciparum/patología , Polimorfismo de Nucleótido Simple , Factor de Necrosis Tumoral alfa/genética , Adulto , Niño , Preescolar , Humanos , Linfotoxina-alfa/inmunología , Linfotoxina beta/inmunología , Malaria Falciparum/complicaciones , Malaria Falciparum/inmunología , Papúa Nueva Guinea , Factor de Necrosis Tumoral alfa/inmunología
16.
J Immunol ; 181(11): 7458-62, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19017933

RESUMEN

Studies in experimental cerebral malaria (ECM) in mice have identified T cells and TNF family members as critical mediators of pathology. In this study we report a role for LIGHT-lymphotoxin beta Receptor (LTbetaR) signaling in the development of ECM and control of parasite growth. Specific blockade of LIGHT-LTbetaR, but not LIGHT-herpesvirus entry mediator interactions, abrogated the accumulation of parasites and the recruitment of pathogenic CD8(+) T cells and monocytes to the brain during infection without affecting early activation of CD4(+) T cells, CD8(+) T cells, or NK cells. Importantly, blockade of LIGHT-LTbetaR signaling caused the expansion of splenic monocytes and an overall enhanced capacity to remove and process Ag during infection, as well as reduced systemic cytokine levels when control mice displayed severe ECM symptoms. In summary, we have discovered a novel pathogenic role for LIGHT and LTbetaR in ECM, identifying this TNF family receptor-ligand interaction as an important immune regulator during experimental malaria.


Asunto(s)
Receptor beta de Linfotoxina/inmunología , Malaria Cerebral/inmunología , Plasmodium berghei/inmunología , Transducción de Señal/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Antígenos de Protozoos/inmunología , Encéfalo/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/genética , Citocinas/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Receptor beta de Linfotoxina/genética , Malaria Cerebral/genética , Ratones , Ratones Noqueados , Monocitos/inmunología , Transducción de Señal/genética , Bazo/inmunología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética
17.
Infect Immun ; 76(7): 3312-20, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18474652

RESUMEN

Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection, predominantly experienced by children and nonimmune adults, which results in significant mortality and long-term sequelae. Previous studies have reported distinct susceptibility gene loci in CBA/CaH (CBA) and C57BL/6 (B6) mice with experimental CM (ECM) caused by infection with Plasmodium berghei ANKA. Here we present an analysis of genome-wide expression profiles in brain tissue taken from B6 and CBA mice with ECM and report significant heterogeneity between the two mouse strains. Upon comparison of the leukocyte composition of ECM brain tissue, microglia were expanded in B6 mice but not CBA mice. Furthermore, circulating levels of gamma interferon, interleukin-10, and interleukin-6 were significantly higher in the serum of B6 mice than in that of CBA mice with ECM. Two therapeutic strategies were applied to B6 and CBA mice, i.e., (i) depletion of regulatory T (Treg) cells prior to infection and (ii) depletion of CD8(+) T cells after the establishment of ECM. Despite the described differences between susceptible mouse strains, depletion of Treg cells before infection attenuated ECM in both B6 and CBA mice. In addition, the depletion of CD8(+) T cells when ECM symptoms are apparent leads to abrogation of ECM in B6 mice and a lack of progression of ECM in CBA mice. These results may have important implications for the development of effective treatments for human CM.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Encéfalo/metabolismo , Susceptibilidad a Enfermedades , Depleción Linfocítica/métodos , Malaria Cerebral/inmunología , Malaria Cerebral/prevención & control , Plasmodium berghei/patogenicidad , Proteínas/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Citocinas/sangre , Perfilación de la Expresión Génica , Malaria Cerebral/parasitología , Malaria Cerebral/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , Análisis de Secuencia de ADN , Especificidad de la Especie , Linfocitos T Reguladores/inmunología
18.
Cell Rep ; 24(6): 1425-1433, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30089254

RESUMEN

IL-1ß requires processing by caspase-1 to generate the active, pro-inflammatory cytokine. Acute IL-1ß secretion from inflammasome-activated macrophages requires caspase-1-dependent GSDMD cleavage, which also induces pyroptosis. Mechanisms of IL-1ß secretion by pyroptotic and non-pyroptotic cells, and the precise functions of caspase-1 and GSDMD therein, are unresolved. Here, we show that, while efficient early secretion of endogenous IL-1ß from primary non-pyroptotic myeloid cells in vitro requires GSDMD, later IL-1ß release in vitro and in vivo proceeds independently of GSDMD. IL-1ß maturation is sufficient for slow, caspase-1/GSDMD-independent secretion of ectopic IL-1ß from resting, non-pyroptotic macrophages, but the speed of IL-1ß release is boosted by inflammasome activation, via caspase-1 and GSDMD. IL-1ß cleavage induces IL-1ß enrichment at PIP2-enriched plasma membrane ruffles, and this is a prerequisite for IL-1ß secretion and is mediated by a polybasic motif within the cytokine. We thus reveal a mechanism in which maturation-induced IL-1ß trafficking facilitates its unconventional secretion.


Asunto(s)
Membrana Celular/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Humanos , Transfección
19.
PLoS One ; 8(2): e57034, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23437303

RESUMEN

Lipopolysaccharide (LPS)-activated macrophages secrete pro-inflammatory cytokines, including tumor necrosis factor (TNF) to elicit innate immune responses. Secretion of these cytokines is also a major contributing factor in chronic inflammatory disease. In previous studies we have begun to elucidate the pathways and molecules that mediate the intracellular trafficking and secretion of TNF. Rab6a and Rab6a' (collectively Rab6) are trans-Golgi-localized GTPases known for roles in maintaining Golgi structure and Golgi-associated trafficking. We found that induction of TNF secretion by LPS promoted the selective increase of Rab6 expression. Depletion of Rab6 (via siRNA and shRNA) resulted in reorganization of the Golgi ribbon into more compact structures that at the resolution of electron microcopy consisted of elongated Golgi stacks that likely arose from fusion of smaller Golgi elements. Concomitantly, the delivery of TNF to the cell surface and subsequent release into the media was reduced. Dominant negative mutants of Rab6 had similar effects in disrupting TNF secretion. In live cells, Rab6-GFP were localized on trans-Golgi network (TGN)-derived tubular carriers demarked by the golgin p230. Rab6 depletion and inactive mutants altered carrier egress and partially reduced p230 membrane association. Our results show that Rab6 acts on TNF trafficking at the level of TGN exit in tubular carriers and our findings suggest Rab6 may stabilize p230 on the tubules to facilitate TNF transport. Both Rab6 isoforms are needed in macrophages for Golgi stack organization and for the efficient post-Golgi transport of TNF. This work provides new insights into Rab6 function and into the role of the Golgi complex in cytokine secretion in inflammatory macrophages.


Asunto(s)
Aparato de Golgi/metabolismo , Macrófagos/metabolismo , Factores de Necrosis Tumoral/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Autoantígenos/metabolismo , Brefeldino A/farmacología , Línea Celular , Membrana Celular/metabolismo , Proteínas de la Matriz de Golgi , Proteínas de la Membrana/metabolismo , Ratones , Mutación , Estabilidad Proteica , Transporte de Proteínas/efectos de los fármacos , Interferencia de ARN , Proteínas de Unión al GTP rab/genética
20.
Endocrinology ; 154(1): 140-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23239817

RESUMEN

We and others have previously demonstrated that congenital deficiency of the nuclear hormone receptor, Rorα1, in staggerer (sg/sg) mice results in resistance to diet-induced obesity and increased insulin sensitivity. Paradoxically, the sg/sg mice are susceptible to atherosclerosis and display impaired innate immunity, underscoring the regulatory links between metabolic disease, inflammation, and susceptibility to infection. Here, we present novel evidence that Rorα1 regulates innate immune function by demonstrating impaired phagocytosis in sg/sg mice. The early stages of Fc-γ receptor-mediated phagocytosis in lipopolysaccharide-activated sg/sg bone marrow-derived macrophages (BMMs) were significantly impaired compared with wild-type cells. Moreover, in sg/sg BMMs, the phagocytic cup membranes had reduced levels of cholesterol. Expression profiling revealed dysregulated expression of genes involved in inflammation and lipid metabolism in sg/sg BMMs. Notably, we identified decreased expression of the mRNA encoding cholesterol 25-hydroxylase (Ch25h), an enzyme that converts cholesterol to 25-hydroxycholesterol (25HC), an oxysterol with emerging roles in immunity. Treatment of sg/sg BMMs with 25HC rescued phagocytosis in a dose-dependent manner, whereas small interfering RNA knockdown of Ch25h mRNA expression in wild-type cells attenuated phagocytosis. Hence, we propose that 25HC is essential for optimizing membrane internalization during phagocytosis and that aberrant Ch25h expression in Rorα1-deficient sg/sg macrophages disrupts phagocytosis. Our studies reveal new roles for Rorα1, Ch25h, and 25HC in phagocytosis. Aberrant 25HC underpins the paradoxical association between insulin sensitivity and impaired innate immunity in Rorα1-deficient mice, heralding a wider and essential role for this oxysterol at the nexus of metabolism and immunity.


Asunto(s)
Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Esteroide Hidroxilasas/metabolismo , Animales , Células Cultivadas , Hidroxicolesteroles/farmacología , Inmunidad Innata/genética , Inmunidad Innata/fisiología , Lipopolisacáridos/farmacología , Masculino , Ratones , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Fagocitosis/genética , Fagocitosis/fisiología , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Receptores de IgG/genética , Receptores de IgG/metabolismo , Esteroide Hidroxilasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA