Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 599(7883): 125-130, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34671159

RESUMEN

Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.


Asunto(s)
Homeostasis , Intestinos/inmunología , Intestinos/fisiología , Neuroglía/inmunología , Neuroglía/fisiología , Regeneración , Adventicia/inmunología , Adventicia/parasitología , Animales , Quimiocina CXCL10/inmunología , Duodeno/inmunología , Duodeno/parasitología , Duodeno/patología , Duodeno/fisiología , Femenino , Gliosis , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Interferón gamma/inmunología , Intestinos/parasitología , Intestinos/patología , Masculino , Ratones , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología
2.
Immunity ; 41(1): 89-103, 2014 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-25035954

RESUMEN

Foxp3(+) T regulatory (Treg) cells prevent inflammatory disease but the mechanistic basis of suppression is not understood completely. Gene silencing by RNA interference can act in a cell-autonomous and non-cell-autonomous manner, providing mechanisms of intercellular regulation. Here, we demonstrate that non-cell-autonomous gene silencing, mediated by miRNA-containing exosomes, is a mechanism employed by Treg cells to suppress T-cell-mediated disease. Treg cells transferred microRNAs (miRNA) to various immune cells, including T helper 1 (Th1) cells, suppressing Th1 cell proliferation and cytokine secretion. Use of Dicer-deficient or Rab27a and Rab27b double-deficient Treg cells to disrupt miRNA biogenesis or the exosomal pathway, respectively, established a requirement for miRNAs and exosomes for Treg-cell-mediated suppression. Transcriptional analysis and miRNA inhibitor studies showed that exosome-mediated transfer of Let-7d from Treg cell to Th1 cells contributed to suppression and prevention of systemic disease. These studies reveal a mechanism of Treg-cell-mediated suppression mediated by miRNA-containing exosomes.


Asunto(s)
Exosomas/genética , MicroARNs/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Animales , Antígenos CD19/inmunología , Linfocitos B/inmunología , Proliferación Celular , Citocinas/metabolismo , ARN Helicasas DEAD-box/genética , Exosomas/inmunología , Exosomas/metabolismo , Femenino , Factores de Transcripción Forkhead/inmunología , Transferencia de Gen Horizontal/genética , Inflamación/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/biosíntesis , MicroARNs/genética , Interferencia de ARN , Ribonucleasa III/genética , Células Th17/inmunología , Proteínas de Unión al GTP rab/genética , Proteínas rab27 de Unión a GTP
3.
Pulm Pharmacol Ther ; 75: 102133, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35644305

RESUMEN

BACKGROUND: Janus Kinases (JAKs) mediate activity of many asthma-relevant cytokines. GDC-0214, an inhaled small molecule JAK1 inhibitor, has previously been shown to reduce fractional exhaled nitric oxide (FeNO) in patients with mild asthma, but required an excessive number of inhalations. AIM: To assess whether GDC-4379, a new inhaled JAK inhibitor, reduces FeNO and peripheral biomarkers of inflammation. METHODS: This study assessed the activity of GDC-4379 in a double-blind, randomized, placebo-controlled, Phase 1 study in patients with mild asthma. Participants included adults (18-65y) with a diagnosis of asthma for ≥6 months, forced expiratory volume in 1 s (FEV1)> 70% predicted, FeNO >40 ppb, using as-needed short-acting beta-agonist medication only. Four sequential, 14-day, ascending-dose cohorts (10 mg QD, 30 mg QD, 40 mg BID, and 80 mg QD) of 12 participants each were randomized 2:1 to GDC-4379 or placebo. The primary activity outcome was percent change from baseline (CFB) in FeNO to Day 14 compared to the pooled placebo group. Safety, tolerability, pharmacokinetics, and pharmacodynamic biomarkers, including blood eosinophils, serum CCL17, and serum CCL18, were also assessed. RESULTS: Of 48 enrolled participants, the mean age was 25 years and 54% were female. Median (range) FeNO at baseline was 79 (41-222) ppb. GDC-4379 treatment led to dose-dependent reductions in FeNO. Compared to placebo, mean (95% CI) percent CFB in FeNO to Day 14 was: -6 (-43, 32) at 10 mg QD, -26 (-53, 2) at 30 mg QD, -55 (-78, -32) at 40 mg BID and -52 (-72, -32) at 80 mg QD. Dose-dependent reductions in blood eosinophils and serum CCL17 were also observed. Higher plasma drug concentrations corresponded with greater FeNO reductions. No serious AEs occurred. The majority of AEs were mild to moderate. The most common AEs were headache and oropharyngeal pain. Minor changes in neutrophils were noted at 80 mg QD, but were not considered clinically meaningful. CONCLUSIONS: In patients with mild asthma, 14-day treatment with GDC-4379 reduced FeNO levels and peripheral biomarkers of inflammation. Treatment was well tolerated without any major safety concerns. AUSTRALIAN NEW ZEALAND CLINICAL TRIALS REGISTRY: ACTRN12619000227190.


Asunto(s)
Asma , Inhibidores de las Cinasas Janus , Adulto , Asma/tratamiento farmacológico , Australia , Biomarcadores , Pruebas Respiratorias , Femenino , Humanos , Inflamación/tratamiento farmacológico , Inhibidores de las Cinasas Janus/efectos adversos , Masculino , Óxido Nítrico
4.
J Allergy Clin Immunol ; 145(2): 666-678.e9, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31445933

RESUMEN

BACKGROUND: Although originally defined as a type 2 (T2) immune-mediated condition, non-T2 cytokines, such as IFN-γ and IL-17A, have been implicated in asthma pathogenesis, particularly in patients with severe disease. IL-10 regulates TH cell phenotypes and can dampen T2 immunity to allergens, but its functions in controlling non-T2 cytokine responses in asthmatic patients are unclear. OBJECTIVE: We sought to determine how IL-10 regulates the balance of TH cell responses to inhaled allergen. METHODS: Allergic airway disease was induced in wild-type, IL-10 reporter, and conditional IL-10 or IL-10 receptor α (IL-10Rα) knockout mice by means of repeated intranasal administration of house dust mite (HDM). IL-10 and IFN-γ signaling were disrupted by using blocking antibodies. RESULTS: Repeated HDM inhalation induced a mixed IL-13/IL-17A response and accumulation of IL-10-producing forkhead box P3-negative effector CD4+ T cells in the lungs. Ablation of T cell-derived IL-10 increased the IFN-γ and IL-17A response to HDM, reducing IL-13 levels and airway eosinophilia without affecting IgE levels or airway hyperresponsiveness. The increased IFN-γ response could be recapitulated by IL-10Rα deletion in CD11c+ myeloid cells or local IL-10Rα blockade. Disruption of the T cell-myeloid IL-10 axis resulted in increased pulmonary monocyte-derived dendritic cell numbers and increased IFN-γ-dependent expression of CXCR3 ligands by airway macrophages, which is suggestive of a feedforward loop of TH1 cell recruitment. Augmented IFN-γ responses in the HDM allergic airway disease model were accompanied by increased disruption of airway epithelium, which was reversed by therapeutic blockade of IFN-γ. CONCLUSIONS: IL-10 from effector T cells signals to CD11c+ myeloid cells to suppress an atypical and pathogenic IFN-γ response to inhaled HDM.


Asunto(s)
Asma/inmunología , Interferón gamma/inmunología , Interleucina-10/inmunología , Células Mieloides/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Alérgenos/inmunología , Animales , Modelos Animales de Enfermedad , Femenino , Hipersensibilidad/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pyroglyphidae/inmunología
5.
J Biol Chem ; 294(41): 15082-15094, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31431507

RESUMEN

Heat shock proteins (Hsps) are highly conserved molecular chaperones that are ubiquitously expressed in all species to aid the solubilization of misfolded proteins, protein degradation, and transport. Elevated levels of Hsp70 have been found in the sputum, serum, and bronchoalveolar lavage (BAL) fluid of asthma patients and are known to correlate with disease severity. However, the function of Hsp70 in allergic airway inflammation has remained largely unknown. This study aimed to determine the role of Hsp70 in airway inflammation and remodeling using a mouse model of allergic airway inflammation. WT and Hsp70 double-knockout (Hsp70.1/.3-/-) mice were sensitized and challenged intratracheally with Schistosoma mansoni soluble egg antigens (SEAs) to induce robust Th2 responses and airway inflammation in the lungs. The lack of Hsp70 resulted in a significant reduction in airway inflammation, goblet cell hyperplasia, and Th2 cytokine production, including IL-4, IL-5, and IL-13. An analysis of the BAL fluid suggested that Hsp70 is critically required for eosinophilic infiltration, collagen accumulation, and Th2 cytokine production in allergic airways. Furthermore, our bone marrow (BM) transfer studies show that SEA-induced airway inflammation, goblet cell hyperplasia, and Th2 cytokine production were attenuated in WT mice that were reconstituted with Hsp70-deficient BM, but these effects were not attenuated in Hsp70-deficient mice that were reconstituted with WT BM. Together, these studies identify a pathogenic role for Hsp70 in hematopoietic cells during allergic airway inflammation; this illustrates the potential utility of targeting Hsp70 to alleviate allergen-induced Th2 cytokines, goblet cell hyperplasia, and airway inflammation.


Asunto(s)
Células Caliciformes/patología , Proteínas HSP70 de Choque Térmico/metabolismo , Hipersensibilidad/metabolismo , Hipersensibilidad/patología , Pulmón/patología , Animales , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Redes Reguladoras de Genes , Hiperplasia/metabolismo , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Pulmón/inmunología , Pulmón/metabolismo , Ratones , Células Th2/inmunología
6.
Nat Immunol ; 9(1): 25-33, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18066066

RESUMEN

The interleukin 4 receptor (IL-4R) is a central mediator of T helper type 2 (T(H)2)-mediated disease and associates with either the common gamma-chain to form the type I IL-4R or with the IL-13R alpha1 chain (IL-13Ralpha1) to form the type II IL-4R. Here we used Il13ra1-/- mice to characterize the distinct functions of type I and type II IL-4 receptors in vivo. In contrast to Il4ra-/- mice, which have weak T(H)2 responses, Il13ra1-/- mice had exacerbated T(H)2 responses. Il13ra1-/- mice showed much less mortality after infection with Schistosoma mansoni and much more susceptibility to Nippostrongylus brasiliensis. IL-13Ralpha1 was essential for allergen-induced airway hyperreactivity and mucus hypersecretion but not for fibroblast or alternative macrophage activation. Thus, type I and II IL-4 receptors exert distinct effects on immune responses.


Asunto(s)
Subunidad alfa1 del Receptor de Interleucina-13/fisiología , Receptores Tipo II de Interleucina-4/fisiología , Células Th2/inmunología , Alérgenos/inmunología , Animales , Antígenos Helmínticos/inmunología , Hiperreactividad Bronquial/inmunología , Células Cultivadas , Susceptibilidad a Enfermedades , Fibroblastos/inmunología , Subunidad alfa1 del Receptor de Interleucina-13/genética , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Moco/metabolismo , Nippostrongylus/fisiología , Schistosoma mansoni/inmunología , Schistosoma mansoni/fisiología , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/mortalidad , Infecciones por Strongylida/inmunología
7.
PLoS Pathog ; 13(7): e1006536, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28759611

RESUMEN

TPL-2 (COT, MAP3K8) kinase activates the MEK1/2-ERK1/2 MAPK signaling pathway in innate immune responses following TLR, TNFR1 and IL-1R stimulation. TPL-2 contributes to type-1/Th17-mediated autoimmunity and control of intracellular pathogens. We recently demonstrated TPL-2 reduces severe airway allergy to house dust mite by negatively regulating type-2 responses. In the present study, we found that TPL-2 deficiency resulted in resistance to Heligmosomoides polygyrus infection, with accelerated worm expulsion, reduced fecal egg burden and reduced worm fitness. Using co-housing experiments, we found resistance to infection in TPL-2 deficient mice (Map3k8-/-) was independent of microbiota alterations in H. polygyrus infected WT and Map3k8-/-mice. Additionally, our data demonstrated immunity to H. polygyrus infection in TPL-2 deficient mice was not due to dysregulated type-2 immune responses. Genome-wide analysis of intestinal tissue from infected TPL-2-deficient mice identified elevated expression of genes involved in chemotaxis and homing of leukocytes and cells, including Ccl24 and alternatively activated genes. Indeed, Map3k8-/-mice had a significant influx of eosinophils, neutrophils, monocytes and Il4GFP+ T cells. Conditional knockout experiments demonstrated that specific deletion of TPL-2 in CD11c+ cells, but not Villin+ epithelial cells, LysM+ myeloid cells or CD4+ T cells, led to accelerated resistance to H. polygyrus. In line with a central role of CD11c+ cells, CD11c+ CD11b+ cells isolated from TPL-2-deficient mice had elevated Ccl24. Finally, Ccl24 neutralization in TPL-2 deficient mice significantly decreased the expression of Arg1, Retnla, Chil3 and Ear11 correlating with a loss of resistance to H. polygyrus. These observations suggest that TPL-2-regulated Ccl24 in CD11c+CD11b+ cells prevents accelerated type-2 mediated immunity to H. polygyrus. Collectively, this study identifies a previously unappreciated role for TPL-2 controlling immune responses to H. polygyrus infection by restricting Ccl24 production.


Asunto(s)
Quimiocina CCL24/inmunología , Quinasas Quinasa Quinasa PAM/inmunología , Nematospiroides dubius/inmunología , Proteínas Proto-Oncogénicas/inmunología , Infecciones por Strongylida/inmunología , Animales , Quimiocina CCL24/genética , Femenino , Humanos , Inmunidad Innata , Quinasas Quinasa Quinasa PAM/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Nematospiroides dubius/genética , Nematospiroides dubius/fisiología , Proteínas Proto-Oncogénicas/genética , Infecciones por Strongylida/enzimología , Infecciones por Strongylida/genética , Infecciones por Strongylida/parasitología , Células Th2/inmunología
8.
J Immunol ; 198(5): 2182-2190, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28100680

RESUMEN

Th22 cells are a major source of IL-22 and have been found at sites of infection and in a range of inflammatory diseases. However, their molecular characteristics and functional roles remain largely unknown because of our inability to generate and isolate pure populations. We developed a novel Th22 differentiation assay and generated dual IL-22/IL-17A reporter mice to isolate and compare pure populations of cultured Th22 and Th17 cells. Il17a fate-mapping and transcriptional profiling provide evidence that these Th22 cells have never expressed IL-17A, suggesting that they are potentially a distinct cell lineage from Th17 cells under in vitro culture conditions. Interestingly, Th22 cells also expressed granzymes, IL-13, and increased levels of Tbet. Using transcription factor-deficient cells, we demonstrate that RORγt and Tbet act as positive and negative regulators of Th22 differentiation, respectively. Furthermore, under Th1 culture conditions in vitro, as well as in an IFN-γ-rich inflammatory environment in vivo, Th22 cells displayed marked plasticity toward IFN-γ production. Th22 cells also displayed plasticity under Th2 conditions in vitro by upregulating IL-13 expression. Our work has identified conditions to generate and characterize Th22 cells in vitro. Further, it provides evidence that Th22 cells develop independently of the Th17 lineage, while demonstrating plasticity toward both Th1- and Th2-type cells.


Asunto(s)
Interleucinas/metabolismo , Proteínas de Dominio T Box/metabolismo , Subgrupos de Linfocitos T/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Plasticidad de la Célula , Células Cultivadas , Humanos , Interleucina-17/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Proteínas de Dominio T Box/genética , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-22
9.
Proc Natl Acad Sci U S A ; 113(5): E568-76, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26787865

RESUMEN

There is a paucity of new therapeutic targets to control allergic reactions and forestall the rising trend of allergic diseases. Although a variety of immune cells contribute to allergy, cytokine-secreting αß(+)CD4(+) T-helper 2 (TH2) cells orchestrate the type-2-driven immune response in a large proportion of atopic asthmatics. To identify previously unidentified putative targets in pathogenic TH2 cells, we performed in silico analyses of recently published transcriptional data from a wide variety of pathogenic TH cells [Okoye IS, et al. (2014) Proc Natl Acad Sci USA 111(30):E3081-E3090] and identified that transcription intermediary factor 1 regulator-alpha (Tif1α)/tripartite motif-containing 24 (Trim24) was predicted to be active in house dust mite (HDM)- and helminth-elicited Il4(gfp+)αß(+)CD4(+) TH2 cells but not in TH1, TH17, or Treg cells. Testing this prediction, we restricted Trim24 deficiency to T cells by using a mixed bone marrow chimera system and found that T-cell-intrinsic Trim24 is essential for HDM-mediated airway allergy and antihelminth immunity. Mechanistically, HDM-elicited Trim24(-/-) T cells have reduced expression of many TH2 cytokines and chemokines and were predicted to have compromised IL-1-regulated signaling. Following this prediction, we found that Trim24(-/-) T cells have reduced IL-1 receptor (IL-1R) expression, are refractory to IL-1ß-mediated activation in vitro and in vivo, and fail to respond to IL-1ß-exacerbated airway allergy. Collectively, these data identify a previously unappreciated Trim24-dependent requirement for IL-1R expression on TH2 cells and an important nonredundant role for T-cell-intrinsic Trim24 in TH2-mediated allergy and antihelminth immunity.


Asunto(s)
Hipersensibilidad/inmunología , Proteínas Nucleares/fisiología , Receptores de Interleucina-1/metabolismo , Células Th2/inmunología , Factores de Transcripción/fisiología , Animales , Helmintos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/genética , Células Th2/metabolismo , Factores de Transcripción/genética
10.
PLoS Pathog ; 12(8): e1005783, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27487182

RESUMEN

Persistent TH2 cytokine responses following chronic helminth infections can often lead to the development of tissue pathology and fibrotic scarring. Despite a good understanding of the cellular mechanisms involved in fibrogenesis, there are very few therapeutic options available, highlighting a significant medical need and gap in our understanding of the molecular mechanisms of TH2-mediated immunopathology. In this study, we found that the Map3 kinase, TPL-2 (Map3k8; Cot) regulated TH2-mediated intestinal, hepatic and pulmonary immunopathology following Schistosoma mansoni infection or S. mansoni egg injection. Elevated inflammation, TH2 cell responses and exacerbated fibrosis in Map3k8-/-mice was observed in mice with myeloid cell-specific (LysM) deletion of Map3k8, but not CD4 cell-specific deletion of Map3k8, indicating that TPL-2 regulated myeloid cell function to limit TH2-mediated immunopathology. Transcriptional and metabolic assays of Map3k8-/-M2 macrophages identified that TPL-2 was required for lipolysis, M2 macrophage activation and the expression of a variety of genes involved in immuno-regulatory and pro-fibrotic pathways. Taken together this study identified that TPL-2 regulated TH2-mediated inflammation by supporting lipolysis and M2 macrophage activation, preventing TH2 cell expansion and downstream immunopathology and fibrosis.


Asunto(s)
Diferenciación Celular/inmunología , Lipólisis/inmunología , Quinasas Quinasa Quinasa PAM/inmunología , Macrófagos/inmunología , Proteínas Proto-Oncogénicas/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular/genética , Fibrosis , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Lipólisis/genética , Quinasas Quinasa Quinasa PAM/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Esquistosomiasis mansoni/genética , Esquistosomiasis mansoni/patología , Células Th2/patología
12.
J Allergy Clin Immunol ; 139(2): 655-666.e7, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27484038

RESUMEN

BACKGROUND: The molecular and cellular pathways driving the pathogenesis of severe asthma are poorly defined. Tumor progression locus 2 (TPL-2) (COT, MAP3K8) kinase activates the MEK1/2-extracellular-signal regulated kinase 1/2 MAP kinase signaling pathway following Toll-like receptor, TNFR1, and IL-1R stimulation. OBJECTIVE: TPL-2 has been widely described as a critical regulator of inflammation, and we sought to investigate the role of TPL-2 in house dust mite (HDM)-mediated allergic airway inflammation. METHODS: A comparative analysis of wild-type and Map3k8-/- mice was conducted. Mixed bone marrow chimeras, conditional knockout mice, and adoptive transfer models were also used. Differential cell counts were performed on the bronchoalveolar lavage fluid, followed by histological analysis of lung sections. Flow cytometry and quantitative PCR was used to measure type 2 cytokines. ELISA was used to assess the production of IgE, type 2 cytokines, and Ccl24. RNA sequencing was used to characterize dendritic cell (DC) transcripts. RESULTS: TPL-2 deficiency led to exacerbated HDM-induced airway allergy, with increased airway and tissue eosinophilia, lung inflammation, and IL-4, IL-5, IL-13, and IgE production. Increased airway allergic responses in Map3k8-/- mice were not due to a cell-intrinsic role for TPL-2 in T cells, B cells, or LysM+ cells but due to a regulatory role for TPL-2 in DCs. TPL-2 inhibited Ccl24 expression in lung DCs, and blockade of Ccl24 prevented the exaggerated airway eosinophilia and lung inflammation in mice given HDM-pulsed Map3k8-/- DCs. CONCLUSIONS: TPL-2 regulates DC-derived Ccl24 production to prevent severe type 2 airway allergy in mice.


Asunto(s)
Asma/inmunología , Quimiocina CCL24/metabolismo , Células Dendríticas/inmunología , Eosinófilos/inmunología , Pulmón/inmunología , Quinasas Quinasa Quinasa PAM/metabolismo , Neumonía/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Animales , Antígenos Dermatofagoides/inmunología , Citocinas/metabolismo , Inmunoglobulina E/sangre , Quinasas Quinasa Quinasa PAM/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Pyroglyphidae/inmunología , Transducción de Señal , Células Th2/inmunología
13.
PLoS Pathog ; 11(7): e1004994, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26147567

RESUMEN

Parasitic helminths establish chronic infections in mammalian hosts. Helminth/Plasmodium co-infections occur frequently in endemic areas. However, it is unclear whether Plasmodium infections compromise anti-helminth immunity, contributing to the chronicity of infection. Immunity to Plasmodium or helminths requires divergent CD4+ T cell-driven responses, dominated by IFNγ or IL-4, respectively. Recent literature has indicated that Th cells, including Th2 cells, have phenotypic plasticity with the ability to produce non-lineage associated cytokines. Whether such plasticity occurs during co-infection is unclear. In this study, we observed reduced anti-helminth Th2 cell responses and compromised anti-helminth immunity during Heligmosomoides polygyrus and Plasmodium chabaudi co-infection. Using newly established triple cytokine reporter mice (Il4gfpIfngyfpIl17aFP635), we demonstrated that Il4gfp+ Th2 cells purified from in vitro cultures or isolated ex vivo from helminth-infected mice up-regulated IFNγ following adoptive transfer into Rag1-/- mice infected with P. chabaudi. Functionally, Th2 cells that up-regulated IFNγ were transcriptionally re-wired and protected recipient mice from high parasitemia. Mechanistically, TCR stimulation and responsiveness to IL-12 and IFNγ, but not type I IFN, was required for optimal IFNγ production by Th2 cells. Finally, blockade of IL-12 and IFNγ during co-infection partially preserved anti-helminth Th2 responses. In summary, this study demonstrates that Th2 cells retain substantial plasticity with the ability to produce IFNγ during Plasmodium infection. Consequently, co-infection with Plasmodium spp. may contribute to the chronicity of helminth infection by reducing anti-helminth Th2 cells and converting them into IFNγ-secreting cells.


Asunto(s)
Coinfección/inmunología , Interferón gamma/metabolismo , Interleucina-12/inmunología , Malaria/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Separación Celular , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interferón gamma/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados , Nematospiroides dubius/inmunología , Plasmodium chabaudi/inmunología , Reacción en Cadena de la Polimerasa
14.
Proc Natl Acad Sci U S A ; 111(30): E3081-90, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25024218

RESUMEN

Allergic diseases, orchestrated by hyperactive CD4(+) Th2 cells, are some of the most common global chronic diseases. Therapeutic intervention relies upon broad-scale corticosteroids with indiscriminate impact. To identify targets in pathogenic Th2 cells, we took a comprehensive approach to identify the microRNA (miRNA) and mRNA transcriptome of highly purified cytokine-expressing Th1, Th2, Th9, Th17, and Treg cells both generated in vitro and isolated ex vivo from allergy, infection, and autoimmune disease models. We report here that distinct regulatory miRNA networks operate to regulate Th2 cells in house dust mite-allergic or helminth-infected animals and in vitro Th2 cells, which are distinguishable from other T cells. We validated several miRNA (miR) candidates (miR-15a, miR-20b, miR-146a, miR-155, and miR-200c), which targeted a suite of dynamically regulated genes in Th2 cells. Through in-depth studies using miR-155(-/-) or miR-146a(-/-) T cells, we identified that T-cell-intrinsic miR-155 was required for type-2 immunity, in part through regulation of S1pr1, whereas T-cell-intrinsic miR-146a was required to prevent overt Th1/Th17 skewing. These data identify miR-155, but not miR-146a, as a potential therapeutic target to alleviate Th2-medited inflammation and allergy.


Asunto(s)
Helmintiasis Animal/inmunología , Hipersensibilidad/inmunología , MicroARNs/inmunología , Células Th2/inmunología , Animales , Perfilación de la Expresión Génica , Helmintiasis Animal/genética , Helmintiasis Animal/patología , Hipersensibilidad/genética , Hipersensibilidad/patología , Ratones , Ratones Noqueados , MicroARNs/genética , Pyroglyphidae/inmunología , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/inmunología , Receptores de Esfingosina-1-Fosfato , Células TH1/inmunología , Células TH1/patología , Células Th17/inmunología , Células Th17/patología , Células Th2/patología
15.
J Immunol ; 192(4): 1778-86, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24442439

RESUMEN

It was reported that host defense against pulmonary Klebsiella pneumoniae infection requires IL-22, which was proposed to be of T cell origin. Supporting a role for IL-22, we found that Il22(-/-) mice had decreased survival compared with wild-type mice after intratracheal infection with K. pneumoniae. Surprisingly, however, Rag2(-/-) mice did not differ from wild-type mice in survival or levels of IL-22 in the lungs postinfection with K. pneumoniae. In contrast, K. pneumoniae-infected Rag2(-/-)Il2rg(-/-) mice failed to produce IL-22. These data suggested a possible role for NK cells or other innate lymphoid cells in host defense and production of IL-22. Unlike NK cell-like innate lymphoid cells that produce IL-22 and display a surface phenotype of NK1.1(-)NKp46(+)CCR6(+), lung NK cells showed the conventional phenotype, NK1.1(+)NKp46(+)CCR6(-). Mice depleted of NK cells using anti-asialo GM1 showed decreased survival and higher lung bacterial counts, as well as increased dissemination of K. pneumoniae to blood and liver, compared with control-treated mice. NK cell depletion also led to decreased production of IL-22 in the lung. Within 1 d postinfection, although there was no increase in the number of lung NK cells, a subset of lung NK cells became competent to produce IL-22, and such cells were found in both wild-type and Rag2(-/-) mice. Our data suggest that, during pulmonary infection of mice with K. pneumoniae, conventional NK cells are required for optimal host defense, which includes the production of IL-22.


Asunto(s)
Interleucinas/metabolismo , Células Asesinas Naturales/inmunología , Infecciones por Klebsiella/inmunología , Neumonía/inmunología , Animales , Antígenos Ly/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Interleucinas/biosíntesis , Interleucinas/genética , Infecciones por Klebsiella/mortalidad , Klebsiella pneumoniae/inmunología , Pulmón/citología , Pulmón/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Subfamilia B de Receptores Similares a Lectina de Células NK/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Neumonía/mortalidad , Receptores CCR6/metabolismo , Sobrevida , Interleucina-22
16.
PLoS Pathog ; 9(6): e1003451, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23825948

RESUMEN

A diverse suite of effector immune responses provide protection against various pathogens. However, the array of effector responses must be immunologically regulated to limit pathogen- and immune-associated damage. CD4(+)Foxp3(+) regulatory T cells (Treg) calibrate immune responses; however, how Treg cells adapt to control different effector responses is unclear. To investigate the molecular mechanism of Treg diversity we used whole genome expression profiling and next generation small RNA sequencing of Treg cells isolated from type-1 or type-2 inflamed tissue following Leishmania major or Schistosoma mansoni infection, respectively. In-silico analyses identified two miRNA "regulatory hubs" miR-10a and miR-182 as critical miRNAs in Th1- or Th2-associated Treg cells, respectively. Functionally and mechanistically, in-vitro and in-vivo systems identified that an IL-12/IFNγ axis regulated miR-10a and its putative transcription factor, Creb. Importantly, reduced miR-10a in Th1-associated Treg cells was critical for Treg function and controlled a suite of genes preventing IFNγ production. In contrast, IL-4 regulated miR-182 and cMaf in Th2-associed Treg cells, which mitigated IL-2 secretion, in part through repression of IL2-promoting genes. Together, this study indicates that CD4(+)Foxp3(+) cells can be shaped by local environmental factors, which orchestrate distinct miRNA pathways preserving Treg stability and suppressor function.


Asunto(s)
Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , MicroARNs/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Linfocitos T Reguladores/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/parasitología , Inflamación/patología , Leishmaniasis Cutánea/genética , Leishmaniasis Cutánea/patología , Ratones , Ratones Noqueados , MicroARNs/genética , Esquistosomiasis mansoni/genética , Esquistosomiasis mansoni/patología , Linfocitos T Reguladores/patología , Células TH1/inmunología , Células TH1/patología , Células Th2/inmunología , Células Th2/patología
17.
J Immunol ; 188(12): 6347-56, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22593615

RESUMEN

The regulators of G protein signaling (RGS) protein superfamily negatively controls G protein-coupled receptor signal transduction pathways. RGS16 is enriched in activated/effector T lymphocytes. In this paper, we show that RGS16 constrains pulmonary inflammation by regulating chemokine-induced T cell trafficking in response to challenge with Schistosoma mansoni. Naive Rgs16(-/-) mice were "primed" for inflammation by accumulation of CCR10(+) T cells in the lung. Upon pathogen exposure, these mice developed more robust granulomatous lung fibrosis than wild-type counterparts. Distinct Th2 or putative Th17 subsets expressing CCR4 or CCR10 accumulated more rapidly in Rgs16(-/-) lungs following challenge and produced proinflammatory cytokines IL-13 and IL-17B. CCR4(+)Rgs16(-/-) Th2 cells migrated excessively to CCL17 and localized aberrantly in challenged lungs. T lymphocytes were partially excluded from lung granulomas in Rgs16(-/-) mice, instead forming peribronchial/perivascular aggregates. Thus, RGS16-mediated confinement of T cells to Schistosome granulomas mitigates widespread cytokine-mediated pulmonary inflammation.


Asunto(s)
Neumonía/inmunología , Proteínas RGS/inmunología , Esquistosomiasis mansoni/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Quimiotaxis de Leucocito/inmunología , Citocinas/biosíntesis , Citometría de Flujo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/metabolismo , Neumonía/microbiología , Proteínas RGS/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Esquistosomiasis mansoni/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo
19.
Expert Opin Drug Discov ; 19(4): 493-506, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38407117

RESUMEN

INTRODUCTION: Inhaled drugs offer advantages for the treatment of respiratory diseases over oral drugs by delivering the drug directly to the lung, thus improving the therapeutic index. There is an unmet medical need for novel therapies for lung diseases, exacerbated by a multitude of challenges for the design of inhaled small molecule drugs. AREAS COVERED: The authors review the challenges and opportunities for the design of inhaled drugs for respiratory diseases with a focus on new target discovery, medicinal chemistry, and pharmacokinetic, pharmacodynamic, and toxicological evaluation of drug candidates. EXPERT OPINION: Inhaled drug discovery is facing multiple unique challenges. Novel biological targets are scarce, as is the guidance for medicinal chemistry teams to design compounds with inhalation-compatible features. It is exceedingly difficult to establish a PK/PD relationship given the complexity of pulmonary PK and the impact of physical properties of the drug substance on PK. PK, PD and toxicology studies are technically challenging and require large amounts of drug substance. Despite the current challenges, the authors foresee that the design of inhaled drugs will be facilitated in the future by our increasing understanding of pathobiology, emerging medicinal chemistry guidelines, advances in drug formulation, PBPK models, and in vitro toxicology assays.


Asunto(s)
Enfermedades Pulmonares , Enfermedades Respiratorias , Humanos , Enfermedades Respiratorias/tratamiento farmacológico , Administración por Inhalación , Enfermedades Pulmonares/tratamiento farmacológico , Descubrimiento de Drogas
20.
PLoS Pathog ; 7(8): e1002171, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21829367

RESUMEN

Despite effective chemotherapy to treat schistosome infections, re-infection rates are extremely high. Resistance to reinfection can develop, however it typically takes several years following numerous rounds of treatment and re-infection, and often develops in only a small cohort of individuals. Using a well-established and highly permissive mouse model, we investigated whether immunoregulatory mechanisms influence the development of resistance. Following Praziquantel (PZQ) treatment of S. mansoni infected mice we observed a significant and mixed anti-worm response, characterized by Th1, Th2 and Th17 responses. Despite the elevated anti-worm response in PBMC's, liver, spleen and mesenteric lymph nodes, this did not confer any protection from a secondary challenge infection. Because a significant increase in IL-10-producing CD4(+)CD44(+)CD25(+)GITR(+) lymphocytes was observed, we hypothesised that IL-10 was obstructing the development of resistance. Blockade of IL-10 combined with PZQ treatment afforded a greater than 50% reduction in parasite establishment during reinfection, compared to PZQ treatment alone, indicating that IL-10 obstructs the development of acquired resistance. Markedly enhanced Th1, Th2 and Th17 responses, worm-specific IgG1, IgG2b and IgE and circulating eosinophils characterized the protection. This study demonstrates that blocking IL-10 signalling during PZQ treatment can facilitate the development of protective immunity and provide a highly effective strategy to protect against reinfection with S. mansoni.


Asunto(s)
Interleucina-10/inmunología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Antihelmínticos/farmacología , Anticuerpos Antihelmínticos/sangre , Anticuerpos Antihelmínticos/inmunología , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Interleucina-10/sangre , Interleucina-10/genética , Ratones , Ratones Transgénicos , Praziquantel/farmacología , Schistosoma mansoni/metabolismo , Esquistosomiasis mansoni/sangre , Esquistosomiasis mansoni/tratamiento farmacológico , Esquistosomiasis mansoni/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células TH1/metabolismo , Células Th17/metabolismo , Células Th2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA