Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Eur J Immunol ; 48(1): 87-98, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28960280

RESUMEN

Dendritic cells (DCs) are essential in dictating the nature and effectiveness of immune responses. In the intestine DCs can be separated into discrete subsets, defined by expression of CD11b and CD103, each with different developmental requirements and distinct functional potential. Recent evidence has shown that different intestinal DC subsets are involved in the induction of T helper (Th)17 and regulatory T cell responses, but the cells that initiate Th2 immune responses are still incompletely understood. We show that in the Th2 response to an intestinal helminth in mice, only CD11b+ and not CD11b- DCs accumulate in the local lymph node, upregulate PDL2 and express markers of alternative activation. An enteric Th1 response instead activated both CD11b+ and CD11b- DCs without eliciting alternative activation in either population. Functionally, only CD11b+ DCs activated during helminth infection supported Th2 differentiation in naive CD4+ T cells. Together our data demonstrate that the ability to prime Th2 cells during intestinal helminth infection, is a selective and inducible characteristic of CD11b+ DCs.


Asunto(s)
Células Dendríticas/inmunología , Activación de Linfocitos/inmunología , Nematospiroides dubius/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Animales , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Células Dendríticas/clasificación , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/parasitología , Intestino Delgado/citología , Intestino Delgado/inmunología , Intestino Delgado/parasitología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Superficie Celular/inmunología , Infecciones por Strongylida/parasitología , Células TH1/inmunología
2.
PLoS Pathog ; 12(9): e1005876, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27598373

RESUMEN

The intestine is a common site for a variety of pathogenic infections. Helminth infections continue to be major causes of disease worldwide, and are a significant burden on health care systems. Lysine methyltransferases are part of a family of novel attractive targets for drug discovery. SETD7 is a member of the Suppressor of variegation 3-9-Enhancer of zeste-Trithorax (SET) domain-containing family of lysine methyltransferases, and has been shown to methylate and alter the function of a wide variety of proteins in vitro. A few of these putative methylation targets have been shown to be important in resistance against pathogens. We therefore sought to study the role of SETD7 during parasitic infections. We find that Setd7-/- mice display increased resistance to infection with the helminth Trichuris muris but not Heligmosomoides polygyrus bakeri. Resistance to T. muris relies on an appropriate type 2 immune response that in turn prompts intestinal epithelial cells (IECs) to alter differentiation and proliferation kinetics. Here we show that SETD7 does not affect immune cell responses during infection. Instead, we found that IEC-specific deletion of Setd7 renders mice resistant to T. muris by controlling IEC turnover, an important aspect of anti-helminth immune responses. We further show that SETD7 controls IEC turnover by modulating developmental signaling pathways such as Hippo/YAP and Wnt/ß-Catenin. We show that the Hippo pathway specifically is relevant during T. muris infection as verteporfin (a YAP inhibitor) treated mice became susceptible to T. muris. We conclude that SETD7 plays an important role in IEC biology during infection.


Asunto(s)
Intestinos/inmunología , Proteína Metiltransferasas/metabolismo , Transducción de Señal , Tricuriasis/inmunología , Trichuris/inmunología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Resistencia a la Enfermedad , Células Epiteliales/parasitología , Células Epiteliales/fisiología , Eliminación de Gen , N-Metiltransferasa de Histona-Lisina , Humanos , Intestinos/parasitología , Intestinos/fisiología , Ratones , Especificidad de Órganos , Fosfoproteínas/metabolismo , Porfirinas/efectos adversos , Proteína Metiltransferasas/genética , Tricuriasis/parasitología , Tricuriasis/patología , Verteporfina , Proteínas Señalizadoras YAP , beta Catenina/metabolismo
3.
J Infect Dis ; 215(8): 1245-1254, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28368463

RESUMEN

Intestinal helminth infections occur predominantly in regions where exposure to enteric bacterial pathogens is also common. Helminth infections inhibit host immunity against microbial pathogens, which has largely been attributed to the induction of regulatory or type 2 (Th2) immune responses. Here we demonstrate an additional 3-way interaction in which helminth infection alters the metabolic environment of the host intestine to enhance bacterial pathogenicity. We show that an ongoing helminth infection increased colonization by Salmonella independently of T regulatory or Th2 cells. Instead, helminth infection altered the metabolic profile of the intestine, which directly enhanced bacterial expression of Salmonella pathogenicity island 1 (SPI-1) genes and increased intracellular invasion. These data reveal a novel mechanism by which a helminth-modified metabolome promotes susceptibility to bacterial coinfection.


Asunto(s)
Coinfección/inmunología , Helmintiasis/inmunología , Parasitosis Intestinales/inmunología , Mucosa Intestinal/metabolismo , Metaboloma , Infecciones por Salmonella/inmunología , Células Th2/inmunología , Animales , Coinfección/microbiología , Coinfección/parasitología , Células HeLa , Humanos , Intestinos/microbiología , Intestinos/parasitología , Ratones , Ratones Endogámicos C57BL , Salmonella typhimurium/genética
4.
J Allergy Clin Immunol ; 135(1): 100-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25145536

RESUMEN

BACKGROUND: Resident gut microbiota are now recognized as potent modifiers of host immune responses in various scenarios. Recently, we demonstrated that perinatal exposure to vancomycin, but not streptomycin, profoundly alters gut microbiota and enhances susceptibility to a TH2 model of allergic asthma. OBJECTIVE: Here we sought to further clarify the etiology of these changes by determining whether perinatal antibiotic treatment has a similar effect on the TH1/TH17-mediated lung disease, hypersensitivity pneumonitis. METHODS: Hypersensitivity pneumonitis was induced in C57BL/6 wild-type or recombination-activating gene 1-deficient mice treated perinatally with vancomycin or streptomycin by repeated intranasal administration of Saccharopolyspora rectivirgula antigen. Disease severity was assessed by measuring lung inflammation, pathology, cytokine responses, and serum antibodies. Microbial community analyses were performed on stool samples via 16S ribosomal RNA pyrosequencing and correlations between disease severity and specific bacterial taxa were identified. RESULTS: Surprisingly, in contrast to our findings in an allergic asthma model, we found that the severity of hypersensitivity pneumonitis was unaffected by vancomycin, but increased dramatically after streptomycin treatment. This likely reflects an effect on the adaptive, rather than innate, immune response because the effects of streptomycin were not observed during the early phases of disease and were abrogated in recombination-activating gene 1-deficient mice. Interestingly, Bacteroidetes dominated the intestinal microbiota of streptomycin-treated animals, while vancomycin promoted the expansion of the Firmicutes. CONCLUSIONS: Perinatal antibiotics exert highly selective effects on resident gut flora, which, in turn, lead to very specific alterations in susceptibility to TH2- or TH1/TH17-driven lung inflammatory disease.


Asunto(s)
Alveolitis Alérgica Extrínseca/inmunología , Alveolitis Alérgica Extrínseca/microbiología , Antibacterianos/efectos adversos , Tracto Gastrointestinal/microbiología , Microbiota , Estreptomicina/efectos adversos , Alveolitis Alérgica Extrínseca/sangre , Alveolitis Alérgica Extrínseca/patología , Animales , Animales Recién Nacidos , Citocinas/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Pulmón/inmunología , Pulmón/patología , Ratones Endogámicos C57BL , Saccharopolyspora , Índice de Severidad de la Enfermedad , Vancomicina/farmacología
5.
Infect Immun ; 83(10): 3881-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26195548

RESUMEN

Foxp3(+) regulatory T (Treg) cells play a key role in suppression of immune responses during parasitic helminth infection, both by controlling damaging immunopathology and by inhibiting protective immunity. During the patent phase of Schistosoma mansoni infection, Foxp3(+) Treg cells are activated and suppress egg-elicited Th2 responses, but little is known of their induction and role during the early prepatent larval stage of infection. We quantified Foxp3(+) Treg cell responses during the first 3 weeks of murine S. mansoni infection in C57BL/6 mice, a time when larval parasites migrate from the skin and transit the lungs en route to the hepatic and mesenteric vasculature. In contrast to other helminth infections, S. mansoni did not elicit a Foxp3(+) Treg cell response during this early phase of infection. We found that the numbers and proportions of Foxp3(+) Treg cells remained unchanged in the lungs, draining lymph nodes, and spleens of infected mice. There was no increase in the activation status of Foxp3(+) Treg cells upon infection as assessed by their expression of CD25, Foxp3, and Helios. Furthermore, infection failed to induce Foxp3(+) Treg cells to produce the suppressive cytokine interleukin 10 (IL-10). Instead, only CD4(+) Foxp3(-) IL-4(+) Th2 cells showed increased IL-10 production upon infection. These data indicate that Foxp3(+) Treg cells do not play a prominent role in regulating immunity to S. mansoni larvae and that the character of the initial immune response invoked by S. mansoni parasites contrasts with the responses to other parasitic helminth infections that promote rapid Foxp3(+) Treg cell responses.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Linfocitos T Reguladores/inmunología , Animales , Movimiento Celular , Femenino , Factores de Transcripción Forkhead/genética , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-4/genética , Interleucina-4/inmunología , Larva/inmunología , Larva/fisiología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Schistosoma mansoni/fisiología , Esquistosomiasis mansoni/genética , Esquistosomiasis mansoni/parasitología , Bazo/inmunología , Linfocitos T Reguladores/parasitología , Células Th2/inmunología
6.
PLoS Pathog ; 9(3): e1003215, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23516361

RESUMEN

The suppression of protective Type 2 immunity is a principal factor driving the chronicity of helminth infections, and has been attributed to a range of Th2 cell-extrinsic immune-regulators. However, the intrinsic fate of parasite-specific Th2 cells within a chronic immune down-regulatory environment, and the resultant impact such fate changes may have on host resistance is unknown. We used IL-4gfp reporter mice to demonstrate that during chronic helminth infection with the filarial nematode Litomosoides sigmodontis, CD4(+) Th2 cells are conditioned towards an intrinsically hypo-responsive phenotype, characterised by a loss of functional ability to proliferate and produce the cytokines IL-4, IL-5 and IL-2. Th2 cell hypo-responsiveness was a key element determining susceptibility to L. sigmodontis infection, and could be reversed in vivo by blockade of PD-1 resulting in long-term recovery of Th2 cell functional quality and enhanced resistance. Contrasting with T cell dysfunction in Type 1 settings, the control of Th2 cell hypo-responsiveness by PD-1 was mediated through PD-L2, and not PD-L1. Thus, intrinsic changes in Th2 cell quality leading to a functionally hypo-responsive phenotype play a key role in determining susceptibility to filarial infection, and the therapeutic manipulation of Th2 cell-intrinsic quality provides a potential avenue for promoting resistance to helminths.


Asunto(s)
Citocinas/metabolismo , Filariasis/inmunología , Filarioidea/inmunología , Células Th2/inmunología , Animales , Antígeno B7-H1/metabolismo , Citocinas/análisis , Susceptibilidad a Enfermedades , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Femenino , Filariasis/parasitología , Citometría de Flujo , Humanos , Activación de Linfocitos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Fenotipo , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células Th2/metabolismo
7.
Cytokine ; 75(1): 51-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25819429

RESUMEN

Type 2 immune responses are defined by the cytokines interleukin 4 (IL-4), IL-5 and IL-13 and the cellular and physiological changes that these cytokines induce, including IgE production, eosinophilia, mast cell degranulation, mucus secretion and smooth muscle contraction. Together these responses provide a "weep and sweep" reflex that is optimised to expel parasitic worms. The same response can also be pathological when mis-timed or activated inappropriately. Current understanding of the orchestration and regulation of type 2 immunity is rapidly advancing, with recent identification of participating innate cells and elucidation of the cytokine signals responsible for their activation. In vivo, the outcome of cytokine signalling is critically dependent on timing, location and context. In this commentary, we describe the spatiotemporal control of type 2 cytokine signalling, consider its implications for bystander cells, and discuss its significance during co-infection.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-4/metabolismo , Transducción de Señal , Animales , Linfocitos B/citología , Efecto Espectador , Citocinas/metabolismo , Humanos , Sistema Inmunológico , Inflamación/metabolismo , Interleucina-13/metabolismo , Pulmón/metabolismo , Ganglios Linfáticos/metabolismo , Ratones , Células Th2/citología
8.
Eur J Immunol ; 43(3): 705-15, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319295

RESUMEN

Foxp3(+) regulatory T (Treg) cells are key immune regulators during helminth infections, and identifying the mechanisms governing their induction is of principal importance for the design of treatments for helminth infections, allergies and autoimmunity. Little is yet known regarding the co-stimulatory environment that favours the development of Foxp3(+) Treg-cell responses during helminth infections. As recent evidence implicates the co-stimulatory receptor ICOS in defining Foxp3(+) Treg-cell functions, we investigated the role of ICOS in helminth-induced Foxp3(+) Treg-cell responses. Infection of ICOS(-/-) mice with Heligmosomoides polygyrus or Schistosoma mansoni led to a reduced expansion and maintenance of Foxp3(+) Treg cells. Moreover, during H. polygyrus infection, ICOS deficiency resulted in increased Foxp3(+) Treg-cell apoptosis, a Foxp3(+) Treg-cell specific impairment in IL-10 production, and a failure to mount putatively adaptive Helios(-) Foxp3(+) Treg-cell responses within the intestinal lamina propria. Impaired lamina propria Foxp3(+) Treg-cell responses were associated with increased production of IL-4 and IL-13 by CD4(+) T cells, demonstrating that ICOS dominantly downregulates Type 2 responses at the infection site, sharply contrasting with its Type 2-promoting effects within lymphoid tissue. Thus, ICOS regulates Type 2 immunity in a tissue-specific manner, and plays a key role in driving Foxp3(+) Treg-cell expansion and function during helminth infections.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Helmintiasis/genética , Helmintiasis/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucina-10/biosíntesis , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Apoptosis/genética , Femenino , Proteína Coestimuladora de Linfocitos T Inducibles/deficiencia , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Masculino , Ratones , Ratones Noqueados , Membrana Mucosa/inmunología , Membrana Mucosa/parasitología , Nematospiroides dubius/inmunología , Células Th2/inmunología
9.
J Immunol ; 187(3): 1411-20, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21705620

RESUMEN

The immune suppression that characterizes human helminth infections can hinder the development of protective immunity or help to reduce pathogenic inflammation. Signaling through the T cell costimulator glucocorticoid-induced TNFR-related protein (GITR) counteracts immune downregulation by augmenting effector T cell responses and abrogating suppression by Foxp3(+) regulatory T cells. Thus, superphysiological Ab-mediated GITR costimulation represents a novel therapy for promoting protective immunity toward parasitic helminths, whereas blocking physiological GITR-GITR ligand (GITRL) interactions may provide a mechanism for dampening pathogenic Th2 inflammation. We investigated the superphysiological and physiological roles of the GITR-GITRL pathway in the development of protective and pathogenic Th2 responses in murine infection models of filariasis (Litomosoides sigmodontis) and schistosomiasis (Schistosoma mansoni). Providing superphysiological GITR costimulation using an agonistic anti-GITR mAb over the first 12 d of L. sigmodontis infection initially increased the quantity of Th2 cells, as well as their ability to produce Th2 cytokines. However, as infection progressed, the Th2 responses reverted to normal infection levels, and parasite killing remained unaffected. Despite the Th2-promoting role of superphysiological GITR costimulation, Ab-mediated blockade of the GITR-GITRL pathway did not affect Th2 cell priming or maintenance during L. sigmodontis infection. Blockade of GITR-GITRL interactions during the acute egg phase of S. mansoni infection resulted in reduced Th2 responses, but this effect was confined to the spleen and did not lead to changes in liver pathology. Thus, although superphysiological GITR costimulation can therapeutically enhance Th2 responses, physiological GITR-GITRL interactions are not required for the development of Th2-mediated resistance or pathology in murine models of filariasis and schistosomiasis.


Asunto(s)
Filarioidea/inmunología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/metabolismo , Activación de Linfocitos/inmunología , Schistosoma mansoni/inmunología , Células Th2/inmunología , Células Th2/parasitología , Factores de Necrosis Tumoral/metabolismo , Animales , Femenino , Filariasis/inmunología , Filariasis/patología , Filariasis/terapia , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Proteína Relacionada con TNFR Inducida por Glucocorticoide/fisiología , Humanos , Inmunidad Innata/genética , Ligandos , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/patología , Esquistosomiasis mansoni/terapia , Células Th2/patología , Factores de Necrosis Tumoral/genética
10.
PLoS One ; 9(2): e88771, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24551160

RESUMEN

IL-7 is a critical cytokine for lymphocyte development. Recent work has highlighted critical roles for IL-7 signaling in mature T cell homeostasis and function, but its role in B cells is less well characterized. Using a knock-in mouse possessing a Tyr to Phe mutation at position 449 (IL-7Rα(449F/449F) mice) within the cytoplasmic SH2-binding motif of IL-7Rα, we evaluated the role of IL-7Rα Y449 motif in spleen B cells. IL-7Rα(449F/449F) mice had reduced numbers and increased death of follicular B cells compared to WT, but had significantly more follicular cells than IL-7Rα(-/-). The death of IL-7Rα(449F/449F) follicular cells was not due to a failure to respond to BAFF or lower levels of BAFF, a critical B cell survival factor. Marginal zone B cells were unaffected by the IL-7Rα(449F/449F) mutation. Any role for TSLP was ruled out, as TSLPR(-/-) mice had an identical B cell phenotype to wild-type mice. Bone marrow chimeras and the absence of IL-7Rα on B cells suggested that IL-7 did not directly regulate mature B cells, but that an IL-7-responsive cell was influencing B cells. IL-7 was also critical at the checkpoint between the T1 and T2 stages in the spleen. IL-7Rα(-/-) mice fail to develop T2 cells, but IL-7Rα(449F/449F) show a reduction compared to WT but not complete absence of T2 cells. We also tested the functional responses of IL-7Rα(449F/449F) to antigens and infection and found no difference in antibody responses to T-dependent or T-independent antigens, or to Influenza/A. IL-7 was important for generation of antibody responses to the intestinal worm H. polygyrus and for naive levels of IgA. Taken together, this suggests that IL-7 regulates follicular B cell numbers and survival in a cell-extrinsic manner, via a bone-marrow derived cell, but is not critical for antibody production outside the gut.


Asunto(s)
Linfocitos B/inmunología , Interleucina-7/inmunología , Receptores de Interleucina-7/inmunología , Transducción de Señal/inmunología , Sustitución de Aminoácidos , Animales , Anticuerpos Antivirales/sangre , Antígenos Virales/sangre , Factor Activador de Células B/genética , Factor Activador de Células B/inmunología , Linfocitos B/citología , Supervivencia Celular , Citocinas/genética , Citocinas/inmunología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Inmunoglobulina G/sangre , Virus de la Influenza A/inmunología , Interleucina-7/genética , Ratones , Ratones Transgénicos , Dominios y Motivos de Interacción de Proteínas , Receptores de Interleucina-7/deficiencia , Receptores de Interleucina-7/genética , Bazo/citología , Bazo/inmunología , Linfopoyetina del Estroma Tímico
11.
Gut Microbes ; 5(4): 522-32, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25144609

RESUMEN

The intestinal microbiota are pivotal in determining the developmental, metabolic and immunological status of the mammalian host. However, the intestinal tract may also accommodate pathogenic organisms, including helminth parasites which are highly prevalent in most tropical countries. Both microbes and helminths must evade or manipulate the host immune system to reside in the intestinal environment, yet whether they influence each other's persistence in the host remains unknown. We now show that abundance of Lactobacillus bacteria correlates positively with infection with the mouse intestinal nematode parasite, Heligmosomoides polygyrus, as well as with heightened regulatory T cell (Treg) and Th17 responses. Moreover, H. polygyrus raises Lactobacillus species abundance in the duodenum of C57BL/6 mice, which are highly susceptible to H. polygyrus infection, but not in BALB/c mice, which are relatively resistant. Sequencing of samples at the bacterial gyrB locus identified the principal Lactobacillus species as L. taiwanensis, a previously characterized rodent commensal. Experimental administration of L. taiwanensis to BALB/c mice elevates regulatory T cell frequencies and results in greater helminth establishment, demonstrating a causal relationship in which commensal bacteria promote infection with an intestinal parasite and implicating a bacterially-induced expansion of Tregs as a mechanism of greater helminth susceptibility. The discovery of this tripartite interaction between host, bacteria and parasite has important implications for both antibiotic and anthelmintic use in endemic human populations.


Asunto(s)
Tracto Gastrointestinal/microbiología , Lactobacillus/fisiología , Interacciones Microbianas , Nematospiroides dubius/fisiología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Tracto Gastrointestinal/inmunología , Interacciones Huésped-Patógeno , Lactobacillus/crecimiento & desarrollo , Lactobacillus/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Nematospiroides dubius/crecimiento & desarrollo , Nematospiroides dubius/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA