Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 16(3): 267-75, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25599562

RESUMEN

The quality of the adaptive immune response depends on the differentiation of distinct CD4(+) helper T cell subsets, and the magnitude of an immune response is controlled by CD4(+)Foxp3(+) regulatory T cells (Treg cells). However, how a tissue- and cell type-specific suppressor program of Treg cells is mechanistically orchestrated has remained largely unexplored. Through the use of Treg cell-specific gene targeting, we found that the suppression of allergic immune responses in the lungs mediated by T helper type 2 (TH2) cells was dependent on the activity of the protein kinase CK2. Genetic ablation of the ß-subunit of CK2 specifically in Treg cells resulted in the proliferation of a hitherto-unexplored ILT3(+) Treg cell subpopulation that was unable to control the maturation of IRF4(+)PD-L2(+) dendritic cells required for the development of TH2 responses in vivo.


Asunto(s)
Quinasa de la Caseína II/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Animales , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular/inmunología , Procesos de Crecimiento Celular/inmunología , Línea Celular , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/inmunología , Humanos , Hipersensibilidad/sangre , Hipersensibilidad/inmunología , Factores Reguladores del Interferón/inmunología , Leucocitos Mononucleares/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Superficie Celular/inmunología , Linfocitos T Reguladores/enzimología , Células Th2/enzimología
2.
PLoS Pathog ; 16(12): e1009121, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33351862

RESUMEN

Parasitic helminths are sensed by the immune system via tissue-derived alarmins that promote the initiation of the appropriate type 2 immune responses. Here we establish the nuclear alarmin cytokine IL-33 as a non-redundant trigger of specifically IL-9-driven and mast cell-mediated immunity to the intestinal parasite Strongyloides ratti. Blockade of endogenous IL-33 using a helminth-derived IL-33 inhibitor elevated intestinal parasite burdens in the context of reduced mast cell activation while stabilization of endogenous IL-33 or application of recombinant IL-33 reciprocally reduced intestinal parasite burdens and increased mast cell activation. Using gene-deficient mice, we show that application of IL-33 triggered rapid mast cell-mediated expulsion of parasites directly in the intestine, independent of the adaptive immune system, basophils, eosinophils or Gr-1+ cells but dependent on functional IL-9 receptor and innate lymphoid cells (ILC). Thereby we connect the described axis of IL-33-mediated ILC2 expansion to the rapid initiation of IL-9-mediated and mast cell-driven intestinal anti-helminth immunity.


Asunto(s)
Interleucina-33/inmunología , Interleucina-9/inmunología , Parasitosis Intestinales/inmunología , Linfocitos/inmunología , Mastocitos/inmunología , Estrongiloidiasis/inmunología , Animales , Inmunidad Innata/inmunología , Intestinos/inmunología , Intestinos/parasitología , Ratones , Strongyloides ratti/inmunología
3.
J Immunol ; 194(4): 1413-6, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595777

RESUMEN

Helminths exploit intrinsic regulatory pathways of the mammalian immune system to dampen the immune response directed against them. In this article, we show that infection with the parasitic nematode Strongyloides ratti induced upregulation of the coinhibitory receptor B and T lymphocyte attenuator (BTLA) predominantly on CD4(+) T cells but also on a small fraction of innate leukocytes. Deficiency of either BTLA or its ligand herpes virus entry mediator (HVEM) resulted in reduced numbers of parasitic adults in the small intestine and reduced larval output throughout infection. Reduced parasite burden in BTLA- and HVEM-deficient mice was accompanied by accelerated degranulation of mucosal mast cells and increased Ag-specific production of the mast cell-activating cytokine IL-9. Our combined results support a model whereby BTLA on CD4(+) T cells and additional innate leukocytes is triggered by HVEM and delivers negative signals into BTLA(+) cells, thereby interfering with the protective immune response to this intestinal parasite.


Asunto(s)
Inmunidad Mucosa/inmunología , Receptores Inmunológicos/inmunología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/inmunología , Transducción de Señal/inmunología , Estrongiloidiasis/inmunología , Animales , Modelos Animales de Enfermedad , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Strongyloides ratti , Linfocitos T/inmunología
4.
Parasitology ; 144(3): 295-315, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-26905057

RESUMEN

The human pathogenic nematode Strongyloides stercoralis infects approximately 30-100 million people worldwide. Analysis of the adaptive immune response to S. stercoralis beyond descriptive studies is challenging, as no murine model for the complete infection cycle is available. However, the combined employment of different models each capable of modelling some features of S. stercoralis life cycle and pathology has advanced our understanding of the immunological mechanisms involved in host defence. Here we review: (i) studies using S. stercoralis third stage larvae implanted in diffusion chambers in the subcutaneous tissue of mice that allow analysis of the immune response to the human pathogenic Strongyloides species; (ii) studies using Strongyloides ratti and Strongyloides venezuelensis that infect mice and rats to extend the analysis to the parasites intestinal life stage and (iii) studies using S. stercoralis infected gerbils to analyse the hyperinfection syndrome, a severe complication of human strongyloidiasis that is not induced by rodent specific Strongyloides spp. We provide an overview of the information accumulated so far showing that Strongyloides spp. elicits a classical Th2 response that culminates in different, site specific, effector functions leading to either entrapment and killing of larvae in the tissues or expulsion of parasitic adults from the intestine.


Asunto(s)
Inmunidad Adaptativa , Enfermedades de los Roedores/inmunología , Strongyloides/inmunología , Strongyloides/patogenicidad , Estrongiloidiasis/veterinaria , Animales , Ratones , Ratas , Enfermedades de los Roedores/parasitología , Estrongiloidiasis/inmunología , Estrongiloidiasis/parasitología , Células Th2/inmunología
5.
Eur J Immunol ; 45(9): 2568-81, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26138667

RESUMEN

Helminth parasites suppress immune responses to prolong their survival within the mammalian host. Thereby not only helminth-specific but also nonhelminth-specific bystander immune responses are suppressed. Here, we use the murine model of Litomosoides sigmodontis infection to elucidate the underlying mechanisms leading to this bystander T-cell suppression. When OT-II T cells specific for the third-party antigen ovalbumin are transferred into helminth-infected mice, these cells respond to antigen-specific stimulation with reduced proliferation compared to activation within non-infected mice. Thus, the presence of parasitic worms in the thoracic cavity translates to suppression of T cells with a different specificity at a different site. By eliminating regulatory receptors, cytokines, and cell populations from this system, we provide evidence for a two-staged process. Parasite products first engage the TGF-ß receptor on host-derived T cells that are central to suppression. In a second step, host-derived T cells produce IL-10 and subsequently suppress the adoptively transferred OT-II T cells. Terminal suppression was IL-10-dependant but independent of intrinsic TGF-ß receptor- or PD-1-mediated signaling in the suppressed OT-II T cells. Blockade of the same key suppression mediators, i.e. TGF-ß- and IL-10 receptor, also ameliorated the suppression of IgG response to bystander antigen vaccination in L. sigmodontis-infected mice.


Asunto(s)
Efecto Espectador/inmunología , Filariasis/inmunología , Interleucina-10/inmunología , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/parasitología , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Filariasis/genética , Filariasis/parasitología , Filariasis/patología , Filarioidea/inmunología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Interleucina-10/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptores de Factores de Crecimiento Transformadores beta/genética , Transducción de Señal , Linfocitos T Reguladores/parasitología , Células Th2/parasitología
6.
PLoS Pathog ; 10(2): e1003913, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24516385

RESUMEN

Accumulating evidence suggests that IL-9-mediated immunity plays a fundamental role in control of intestinal nematode infection. Here we report a different impact of Foxp3⁺ regulatory T cells (Treg) in nematode-induced evasion of IL-9-mediated immunity in BALB/c and C57BL/6 mice. Infection with Strongyloides ratti induced Treg expansion with similar kinetics and phenotype in both strains. Strikingly, Treg depletion reduced parasite burden selectively in BALB/c but not in C57BL/6 mice. Treg function was apparent in both strains as Treg depletion increased nematode-specific humoral and cellular Th2 response in BALB/c and C57BL/6 mice to the same extent. Improved resistance in Treg-depleted BALB/c mice was accompanied by increased production of IL-9 and accelerated degranulation of mast cells. In contrast, IL-9 production was not significantly elevated and kinetics of mast cell degranulation were unaffected by Treg depletion in C57BL/6 mice. By in vivo neutralization, we demonstrate that increased IL-9 production during the first days of infection caused accelerated mast cell degranulation and rapid expulsion of S. ratti adults from the small intestine of Treg-depleted BALB/c mice. In genetically mast cell-deficient (Cpa3-Cre) BALB/c mice, Treg depletion still resulted in increased IL-9 production but resistance to S. ratti infection was lost, suggesting that IL-9-driven mast cell activation mediated accelerated expulsion of S. ratti in Treg-depleted BALB/c mice. This IL-9-driven mast cell degranulation is a central mechanism of S. ratti expulsion in both, BALB/c and C57BL/6 mice, because IL-9 injection reduced and IL-9 neutralization increased parasite burden in the presence of Treg in both strains. Therefore our results suggest that Foxp3⁺ Treg suppress sufficient IL-9 production for subsequent mast cell degranulation during S. ratti infection in a non-redundant manner in BALB/c mice, whereas additional regulatory pathways are functional in Treg-depleted C57BL/6 mice.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Interleucina-9/inmunología , Mastocitos/inmunología , Estrongiloidiasis/inmunología , Linfocitos T Reguladores/inmunología , Animales , Degranulación de la Célula/inmunología , Ensayo de Inmunoadsorción Enzimática , Interleucina-9/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Strongyloides ratti/inmunología , Estrongiloidiasis/metabolismo , Subgrupos de Linfocitos T/inmunología
7.
Eur J Immunol ; 43(7): 1799-805, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23529858

RESUMEN

IL-10, a cytokine with pleiotropic functions is produced by many different cells. Although IL-10 may be crucial for initiating protective Th2 responses to helminth infection, it may also function as a suppressive cytokine preventing immune pathology or even contributing to helminth-induced immune evasion. Here, we show that B cells and T cells produce IL-10 during murine Litomosoides sigmodontis infection. IL-10-deficient mice produced increased amounts of L. sigmodontis-specific IFN-γ and IL-13 suggesting a suppressive role for IL-10 in the initiation of the T-cell response to infection. Using cell type-specific IL-10-deficient mice, we dissected different functions of T-cell- and B-cell-derived IL-10. Litomosoides sigmodontis-specific IFN-γ, IL-5, and IL-13 production increased in the absence of T-cell-derived IL-10 at early and late time points of infection. In contrast, B-cell-specific IL-10 deficiency did not lead to significant changes in L. sigmodontis-specific cytokine production compared to WT mice. Our results suggest that the initiation of Ag-specific cellular responses during L. sigmodontis infection is suppressed by T-cell-derived IL-10 and not by B-cell-derived IL-10.


Asunto(s)
Linfocitos B/inmunología , Filariasis/inmunología , Filarioidea/inmunología , Interleucina-10/inmunología , Linfocitos T/inmunología , Animales , Linfocitos B/metabolismo , Modelos Animales de Enfermedad , Citometría de Flujo , Interleucina-10/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/metabolismo
8.
J Neuroinflammation ; 11: 86, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24886419

RESUMEN

BACKGROUND: Increasing evidence suggests that inflammation associated with microglial cell activation in the substantia nigra (SN) of patients with Parkinson disease (PD) is not only a consequence of neuronal degeneration, but may actively sustain dopaminergic (DA) cell loss over time. We aimed to study whether the intracellular chaperone heat shock protein 60 (Hsp60) could serve as a signal of CNS injury for activation of microglial cells. METHODS: Hsp60 mRNA expression in the mesencephalon and the striatum of C57/BL6 mice treated with MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) and the Hsp60/TH mRNA ratios in the SN of PD patients and aged-matched subjects were measured. To further investigate a possible link between the neuronal Hsp60 response and PD-related cellular stress, Hsp60 immunoblot analysis and quantification in cell lysates from SH-SY5Y after treatment with 100 µM MPP+ (1-methyl-4-phenylpyridinium) at different time points (6, 12, 24 and 48 hours) compared to control cells were performed. Additional MTT and LDH assay were used. We next addressed the question as to whether Hsp60 influences the survival of TH+ neurons in mesencephalic neuron-glia cultures treated either with MPP+ (1 µM), hHsp60 (10 µg/ml) or a combination of both. Finally, we measured IL-1ß, IL-6, TNF-α and NO-release by ELISA in primary microglial cell cultures following treatment with different hHsp60 preparations. Control cultures were exposed to LPS. RESULTS: In the mesencephalon and striatum of mice treated with MPTP and also in the SN of PD patients, we found that Hsp60 mRNA was up-regulated. MPP+, the active metabolite of MPTP, also caused an increased expression and release of Hsp60 in the human dopaminergic cell line SH-SY5Y. Interestingly, in addition to being toxic to DA neurons in primary mesencephalic cultures, exogenous Hsp60 aggravated the effects of MPP+. Yet, although we demonstrated that Hsp60 specifically binds to microglial cells, it failed to stimulate the production of pro-inflammatory cytokines or NO by these cells. CONCLUSIONS: Overall, our data suggest that Hsp60 is likely to participate in DA cell death in PD but via a mechanism unrelated to cytokine release.


Asunto(s)
Chaperonina 60/metabolismo , Cuerpo Estriado/patología , Neuronas Dopaminérgicas/metabolismo , Intoxicación por MPTP/patología , Mesencéfalo/patología , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Chaperonina 60/genética , Modelos Animales de Enfermedad , Dopaminérgicos/farmacología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Óxido Nítrico/metabolismo , Unión Proteica/efectos de los fármacos , ARN Mensajero/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
9.
Philos Trans R Soc Lond B Biol Sci ; 379(1894): 20230004, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38008122

RESUMEN

The Strongyloides genus of parasitic nematodes have a fascinating life cycle and biology, but are also important pathogens of people and a World Health Organization-defined neglected tropical disease. Here, a community of Strongyloides researchers have posed thirteen major questions about Strongyloides biology and infection that sets a Strongyloides research agenda for the future. This article is part of the Theo Murphy meeting issue 'Strongyloides: omics to worm-free populations'.


Asunto(s)
Estadios del Ciclo de Vida , Strongyloides , Animales , Humanos
10.
Eur J Immunol ; 42(4): 890-900, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22161305

RESUMEN

Malaria is still responsible for up to 1 million deaths per year worldwide, highlighting the need for protective malaria vaccines. Helminth infections that are prevalent in malaria endemic areas can modulate immune responses of the host. Here we show that Strongy-Ioides ratti, a gut-dwelling nematode that causes transient infections, did not change the efficacy of vaccination against Plasmodium berghei. An ongoing infection with Litomosoides sigmodontis, a tissue-dwelling filaria that induces chronic infections in BALB/c mice, significantly interfered with vaccination efficacy. The induction of P. berghei circumspor-ozoite protein (CSP)-specific CD8(+) T cells, achieved by a single immunization with a CSP fusion protein, was diminished in L. sigmodontis-infected mice. This modulation was reflected by reduced frequencies of CSP-specific CD8(+) T cells, reduced CSP-specific IFN-y and TNF-a production, reduced CSP-specific cytotoxicity, and reduced protection against P. berghei challenge infection. Implementation of a more potent vaccine regime, by first priming with CSP-expressing recombinant live Salmonella prior to CSP fusion protein immunization, restored induction of CSP-specific CD8(+) T cells and conferred almost sterile immunity to P. berghei challenge infection also in L. sigmodontis-infected mice. In summary, we show that appropriate vaccination regimes can overcome helminth-induced interference with vaccination efficacy.


Asunto(s)
Antígenos de Protozoos/farmacología , Linfocitos T CD8-positivos/inmunología , Filariasis/inmunología , Filarioidea/inmunología , Malaria/inmunología , Plasmodium berghei/inmunología , Proteínas Protozoarias/farmacología , Strongyloides ratti/inmunología , Estrongiloidiasis/inmunología , Animales , Antígenos de Protozoos/inmunología , Inmunización , Interferón gamma/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Protozoarias/inmunología , Ratas , Ratas Wistar , Salmonella/inmunología , Sigmodontinae , Factor de Necrosis Tumoral alfa/inmunología
11.
J Immunol ; 187(8): 4088-99, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21900178

RESUMEN

One third of the human population is infected with helminth parasites. To promote their longevity and to limit pathology, helminths have developed several strategies to suppress the immune response of their host. As this immune suppression also acts on unrelated third-party Ags, a preexisting helminth infection may interfere with vaccination efficacy. In this study, we show that natural infection with Litomosoides sigmodontis suppressed the humoral response to thymus-dependent but not to thymus-independent model Ags in C57BL/6 mice. Thereby, we provide evidence that reduced humoral responses were mediated by interference with Th cell function rather than by direct suppression of B cells in L. sigmodontis-infected mice. We directly demonstrate suppression of Ag-specific proliferation in OVA-specific Th cells after adoptive transfer into L. sigmodontis-infected mice that led to equally reduced production of OVA-specific IgG. Transferred Th cells displayed increased frequencies of Foxp3(+) after in vivo stimulation within infected but not within naive mice. Helminth-mediated suppression was induced by established L. sigmodontis infections but was completely independent of the individual worm burden. Using DEREG mice, we rule out a central role for host-derived regulatory T cells in the suppression of transferred Th cell proliferation. In contrast, we show that L. sigmodontis-induced, host-derived IL-10 mediated Foxp3 induction in transferred Th cells and significantly contributed to the observed Th cell hypoproliferation within infected mice.


Asunto(s)
Antígenos/inmunología , Filariasis/inmunología , Inmunidad Humoral/inmunología , Interleucina-10/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Filarioidea/inmunología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
12.
J Immunol ; 186(7): 4295-305, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21335490

RESUMEN

To escape expulsion by their host's immune system, pathogenic nematodes exploit regulatory pathways that are intrinsic parts of the mammalian immune system, such as regulatory T cells (Tregs). Using depletion of Treg mice, we showed that Foxp3(+) Treg numbers increased rapidly during infection with the nematode Strongyloides ratti. Transient depletion of Tregs during the first days of infection led to dramatically reduced worm burden and larval output, without aggravation of immune pathology. The transient absence of Tregs during primary infection did not interfere with the generation of protective memory. Depletion of Tregs at later time points of infection (i.e., day 4) did not improve resistance, suggesting that Tregs exert their counterregulatory function during the priming of S. ratti-specific immune responses. Improved resistance upon early Treg depletion was accompanied by accelerated and prolonged mast cell activation and increased production of types 1 and 2 cytokines. In contrast, the blockade of the regulatory receptor CTLA-4 specifically increased nematode-specific type 2 cytokine production. Despite this improved immune response, resistance to the infection was only marginally improved. Taken together, we provide evidence that Treg expansion during S. ratti infection suppresses the protective immune response to this pathogenic nematode and, thus, represents a mechanism of immune evasion.


Asunto(s)
Diferenciación Celular/inmunología , Factores de Transcripción Forkhead/biosíntesis , Strongyloides ratti/inmunología , Estrongiloidiasis/inmunología , Estrongiloidiasis/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Animales , Antígenos CD/inmunología , Antígeno CTLA-4 , Diferenciación Celular/genética , Células Cultivadas , Evasión Inmune/genética , Inmunidad Innata/genética , Memoria Inmunológica/genética , Depleción Linfocítica , Linfopenia/genética , Linfopenia/inmunología , Linfopenia/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ratas , Ratas Wistar , Estrongiloidiasis/parasitología , Linfocitos T Reguladores/parasitología
13.
Parasitol Res ; 112(9): 3335-46, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23820606

RESUMEN

The macrophage migration inhibitory factors (MIFs) from the filarial parasite Onchocerca volvulus (OvMIF) were compared to the MIFs from the free-living nematode Caenorhabditis elegans (CeMIF) with respect to molecular, biochemical and immunological properties. Except for CeMIF-4, all other MIFs demonstrated tautomerase activity. Surprisingly, OvMIF-1 displayed oxidoreductase activity. The strongest immunostaining for OvMIF-1 was observed in the outer cellular covering of the adult worm body, the syncytial hypodermis; moderate immunostaining was observed in the uterine wall. The generation of a strong humoral immune response towards OvMIF-1 and reduced reactivity to OvMIF-2 was indicated by high IgG levels in patients infected with O. volvulus and cows infected with the closely related Onchocerca ochengi, both MIFs revealing identical amino acid sequences. Using Litomosoides sigmodontis-infected mice, a laboratory model for filarial infection, MIFs derived from the tissue-dwelling O. volvulus, the rodent gut-dwelling Strongyloides ratti and from free-living C. elegans were recognized, suggesting that L. sigmodontis MIF-specific IgM and IgG1 were produced during L. sigmodontis infection of mice and cross-reacted with all MIF proteins tested. Thus, MIF apparently functions as a target of B cell response during nematode infection, but in the natural Onchocerca-specific human and bovine infection, the induced antibodies can discriminate between MIFs derived from parasitic or free-living nematodes.


Asunto(s)
Caenorhabditis elegans/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Onchocerca volvulus/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Antihelmínticos/biosíntesis , Caenorhabditis elegans/genética , Caenorhabditis elegans/inmunología , Bovinos , Reacciones Cruzadas , Femenino , Filariasis/inmunología , Filariasis/parasitología , Filarioidea/inmunología , Filarioidea/fisiología , Humanos , Inmunidad Humoral , Factores Inhibidores de la Migración de Macrófagos/genética , Factores Inhibidores de la Migración de Macrófagos/inmunología , Factores Inhibidores de la Migración de Macrófagos/aislamiento & purificación , Masculino , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Onchocerca volvulus/genética , Onchocerca volvulus/inmunología , Oncocercosis/inmunología , Oncocercosis/parasitología , Proteínas Recombinantes , Alineación de Secuencia , Análisis de Secuencia de ADN , Sigmodontinae , Especificidad por Sustrato
14.
Immunol Lett ; 255: 62-66, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36889363

RESUMEN

Helminth parasites infect more than a quarter of the human population and inflict significant changes to the immunological status of their hosts. Several human studies report impaired responses to vaccinations in helminth-infected individuals. Analysing the impact of helminth infections on the efficacy of influenza vaccinations in the mouse system helps to elucidate the underlying immunological processes. Concurrent infection with the parasitic nematode Litomosoides sigmodontis reduced the quantity and quality of antibody responses to vaccination against seasonal influenza in BALB/c and C57BL/6 mice. This led to impaired vaccination-induced protection against challenge infections with the human pathogenic 2009 pandemic H1N1 influenza A virus in helminth-infected mice. Impaired responses were also observed if vaccinations were performed after immune-driven or drug-induced clearance of a previous helminth infection. Mechanistically, the suppression was associated with a systemic and sustained expansion of IL-10-producing CD4+CD49b+LAG-3+ type 1 regulatory T cells and partially abrogated by in vivo blockade of the IL-10 receptor. In summary, these findings raise the concern that individuals in helminth-endemic areas may not always benefit from vaccinations, even in the absence of an acute and diagnosable helminth infection.


Asunto(s)
Filariasis , Filarioidea , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana , Humanos , Ratones , Animales , Filariasis/parasitología , Filariasis/patología , Ratones Endogámicos C57BL , Vacunación , Ratones Endogámicos BALB C
15.
Front Immunol ; 14: 1182502, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37469513

RESUMEN

The transcription factor Interferon Regulatory Factor 4 (IRF4) is central in control of T cell activation and differentiation. Deficiency of IRF4 results in severe immune deficiency and affects maturation and function of most if not all T cell subsets. Here we use mouse infection models for Citrobacter rodentium and Strongyloides ratti to analyze the function of IRF4 in T helper (Th) 17 and Th2 cell responses, respectively. IRF4 deficient mice were impaired in the control of both pathogens, failed to mount Th17 and Th2 cell responses and showed impaired recruitment of T helper cells to the intestine, the infection site of both pathogens. Compromised intestinal migration was associated with reduced expression of the intestinal homing receptors α4ß7 integrin, CCR9 and GPR15. Identification of IRF4 binding sites in the gene loci of these receptors suggests a direct control of their expression by IRF4. Competitive T cell transfer assays further demonstrated that loss of one functional Irf4 allele already affected intestinal accumulation and Th2 and Th17 cell generation, indicating that lower IRF4 levels are of disadvantage for Th2 and Th17 cell differentiation as well as their migration to the intestine. Conversion of peripheral CD4+ T cells from an Irf4 wildtype to an Irf4 heterozygous or from an Irf4 heterozygous to a homozygous mutant genotype after C. rodentium or S. ratti infection did not reduce their capacity to produce Th17 or Th2 cytokines and only partially affected their persistence in the intestine, revealing that IRF4 is not essential for maintenance of the Th2 and Th17 phenotype and for survival of these T helper cells in the intestine. In conclusion, we demonstrate that the expression levels of IRF4 determine Th2 and Th17 cell differentiation and their intestinal accumulation but that IRF4 expression is not crucial for Th2 and Th17 cell survival.


Asunto(s)
Linfocitos T CD4-Positivos , Movimiento Celular , Factores Reguladores del Interferón , Intestinos , Animales , Ratones , Regulación de la Expresión Génica , Factores Reguladores del Interferón/metabolismo , Células Th17 , Células Th2 , Linfocitos T CD4-Positivos/citología
16.
J Exp Med ; 220(12)2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37773047

RESUMEN

Adaptation of immune cells to tissue-specific microenvironments is a crucial process in homeostasis and inflammation. Here, we show that murine effector type 2 innate lymphoid cells (ILC2s) from various organs are equally effective in repopulating ILC2 niches in other anatomical locations where they adapt tissue-specific phenotypes of target organs. Single-cell transcriptomics of ILC2 populations revealed upregulation of retinoic acid (RA) signaling in ILC2s during adaptation to the small intestinal microenvironment, and RA signaling mediated reprogramming of kidney effector ILC2s toward the small intestinal phenotype in vitro and in vivo. Inhibition of intestinal ILC2 adaptation by blocking RA signaling impaired worm expulsion during Strongyloides ratti infection, indicating functional importance of ILC2 tissue imprinting. In conclusion, this study highlights that effector ILC2s retain the ability to adapt to changing tissue-specific microenvironments, enabling them to exert tissue-specific functions, such as promoting control of intestinal helminth infections.


Asunto(s)
Inmunidad Innata , Tretinoina , Ratones , Animales , Tretinoina/farmacología , Linfocitos , Intestinos , Inflamación , Citocinas
17.
Exp Parasitol ; 132(1): 69-75, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21723282

RESUMEN

It is estimated that over one third of the world population is infected with helminths, Strongyloides ssp. accounting for approximately 30-100 million cases. As helminth infections often result in a modulation of the host's immune system, infected people may display impaired responses to concurrent infections and to third party antigens. Here, we employ the experimental system of murine Strongyloides ratti infection to investigate the impact of helminth infections on experimental vaccinations. We demonstrate that concurrent infection with S. ratti strongly affected the humoral response to a thymus dependent model antigen, whereby predominantly Th1 associated IgG2b production was suppressed. We provide evidence that this suppression was due to modulation of T helper cell and not B cell function as the responses to a thymus independent model antigen remained unchanged in S. ratti infected mice. Moreover, using an adoptive transfer system, we show that infection with S. ratti directly interfered with antigen-specific proliferation of T cell receptor transgenic CD4(+) T helper cells in vivo. Finally, using IL-10 deficient mice and mice that selectively lack T helper cell derived IL-10 we rule out a role for host-derived IL-10 in mediating the suppression of thymus dependent model antigen response in S. ratti infected mice.


Asunto(s)
Antígenos Helmínticos/inmunología , Linfocitos B/inmunología , Strongyloides ratti/inmunología , Estrongiloidiasis/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Traslado Adoptivo , Animales , Femenino , Inmunización , Inmunoglobulina G/biosíntesis , Interleucina-10/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ratas , Ratas Wistar , Timo/inmunología
18.
PLoS One ; 17(3): e0266456, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35358281

RESUMEN

The world health organization estimates that more than a quarter of the human population is infected with parasitic worms that are called helminths. Many helminths suppress the immune system of their hosts to prolong their survival. This helminth-induced immunosuppression "spills over" to unrelated antigens and can suppress the immune response to vaccination against other pathogens. Indeed, several human studies have reported a negative correlation between helminth infections and responses to vaccinations. Using mice that are infected with the parasitic nematode Litomosoides sigmodontis as a model for chronic human filarial infections, we reported previously that concurrent helminth infection impaired the vaccination-induced protection against the human pathogenic 2009 pandemic H1N1 influenza A virus (2009 pH1N1). Vaccinated, helminth-infected mice produced less neutralizing, influenza-specific antibodies than vaccinated naïve control mice. Consequently helminth-infected and vaccinated mice were not protected against a challenge infection with influenza virus but displayed high virus burden in the lung and a transient weight loss. In the current study we tried to improve the vaccination efficacy using vaccines that are licensed for humans. We either introduced a prime-boost vaccination regimen using the non-adjuvanted anti-influenza vaccine Begripal or employed the adjuvanted influenza vaccine Fluad. Although both strategies elevated the production of influenza-specific antibodies and protected mice from the transient weight loss that is caused by an influenza challenge infection, sterile immunity was not achieved. Helminth-infected vaccinated mice still had high virus burden in the lung while non-helminth-infected vaccinated mice rapidly cleared the virus. In summary we demonstrate that basic improvements of influenza vaccination regimen are not sufficient to confer sterile immunity on the background of helminth-induced immunosuppression, despite amelioration of pathology i.e. weight loss. Our findings highlight the risk of failed vaccinations in helminth-endemic areas, especially in light of the ongoing vaccination campaign to control the COVID-19 pandemic.


Asunto(s)
COVID-19 , Helmintiasis , Helmintos , Subtipo H1N1 del Virus de la Influenza A , Virus de la Influenza A , Vacunas contra la Influenza , Gripe Humana , Infecciones por Orthomyxoviridae , Adyuvantes Inmunológicos , Animales , Anticuerpos Antivirales , Humanos , Gripe Humana/complicaciones , Gripe Humana/prevención & control , Ratones , Pandemias , Vacunación , Pérdida de Peso
20.
J Immunol ; 182(5): 2827-34, 2009 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-19234177

RESUMEN

The transmembrane glycoprotein CD83 is an important regulator of both thymic T cell maturation and peripheral T cell response. Recent studies suggested that CD83 is also involved in the regulation of B cell maturation, activation, and homeostasis. In this study, we show that in vivo overexpression of CD83 dose dependently interfered with the Ig response to thymus-dependent and thymus-independent model Ag immunization. CD83 deficiency, in contrast, which was restricted to B cells in mixed bone marrow chimeras, led to unchanged or even slightly increased Ig responses. Strikingly, the engagement of CD83 that is naturally up-regulated on wild-type B cells by injection of anti-CD83 mAb in vivo induced a 100-fold increase in the IgG1 response to immunization. Kinetic analysis revealed that CD83 had to be engaged simultaneously or shortly after the B cell activation through injection of Ag, to modulate the IgG1 secretion. Furthermore, using mixed bone marrow chimeras in which either selectively the B cells or the dendritic cells were CD83 deficient, we demonstrate that anti-CD83 mAb mediated its biologic effect by engaging CD83 on B cells and not on CD11c(+) dendritic cells. Taken together, we provide strong evidence that CD83 transduces regulatory signals into the very B cell on which it is expressed.


Asunto(s)
Antígenos CD/inmunología , Antígenos CD/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Inmunoglobulinas/inmunología , Inmunoglobulinas/metabolismo , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Animales , Formación de Anticuerpos/genética , Antígenos CD/biosíntesis , Relación Dosis-Respuesta Inmunológica , Ficoll/administración & dosificación , Ficoll/inmunología , Haptenos/administración & dosificación , Haptenos/inmunología , Hemocianinas/administración & dosificación , Hemocianinas/inmunología , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/sangre , Inmunoglobulina M/biosíntesis , Inmunoglobulina M/sangre , Inmunoglobulinas/biosíntesis , Inmunoglobulinas/deficiencia , Terapia de Inmunosupresión , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Nitrofenoles/administración & dosificación , Nitrofenoles/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo , Antígeno CD83
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA