Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Science ; 384(6692): 159-160, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38574173

RESUMO

Androgen signaling skews skin immunity toward reduced inflammation in male mice.


Assuntos
Androgênios , Caracteres Sexuais , Masculino , Feminino , Camundongos , Animais , Imunidade Inata , Linfócitos , Células Dendríticas
2.
J Clin Invest ; 134(6)2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38487999

RESUMO

Allergic asthma generally starts during early life and is linked to substantial tissue remodeling and lung dysfunction. Although angiogenesis is a feature of the disrupted airway, the impact of allergic asthma on the pulmonary microcirculation during early life is unknown. Here, using quantitative imaging in precision-cut lung slices (PCLSs), we report that exposure of neonatal mice to house dust mite (HDM) extract disrupts endothelial cell/pericyte interactions in adventitial areas. Central to the blood vessel structure, the loss of pericyte coverage was driven by mast cell (MC) proteases, such as tryptase, that can induce pericyte retraction and loss of the critical adhesion molecule N-cadherin. Furthermore, spatial transcriptomics of pediatric asthmatic endobronchial biopsies suggests intense vascular stress and remodeling linked with increased expression of MC activation pathways in regions enriched in blood vessels. These data provide previously unappreciated insights into the pathophysiology of allergic asthma with potential long-term vascular defects.


Assuntos
Asma , Mastócitos , Humanos , Criança , Animais , Camundongos , Mastócitos/patologia , Pericitos/metabolismo , Células Endoteliais/metabolismo , Asma/patologia , Pulmão/patologia , Alérgenos , Pyroglyphidae , Modelos Animais de Doenças
3.
Nat Commun ; 14(1): 6039, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37758700

RESUMO

Aberrant expansion of KRT5+ basal cells in the distal lung accompanies progressive alveolar epithelial cell loss and tissue remodelling during fibrogenesis in idiopathic pulmonary fibrosis (IPF). The mechanisms determining activity of KRT5+ cells in IPF have not been delineated. Here, we reveal a potential mechanism by which KRT5+ cells migrate within the fibrotic lung, navigating regional differences in collagen topography. In vitro, KRT5+ cell migratory characteristics and expression of remodelling genes are modulated by extracellular matrix (ECM) composition and organisation. Mass spectrometry- based proteomics revealed compositional differences in ECM components secreted by primary human lung fibroblasts (HLF) from IPF patients compared to controls. Over-expression of ECM glycoprotein, Secreted Protein Acidic and Cysteine Rich (SPARC) in the IPF HLF matrix restricts KRT5+ cell migration in vitro. Together, our findings demonstrate how changes to the ECM in IPF directly influence KRT5+ cell behaviour and function contributing to remodelling events in the fibrotic niche.


Assuntos
Fibrose Pulmonar Idiopática , Humanos , Matriz Extracelular , Células Epiteliais Alveolares , Transporte Biológico , Movimento Celular , Queratina-5
4.
Immunity ; 56(2): 229-231, 2023 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-36792567

RESUMO

Communication between nerves and group 2 innate lymphoid cells (ILC2s) is thought to regulate allergic airway inflammation, but the molecular mechanisms are unclear. In this issue of Immunity, Cao et al. uncover an essential role for dopamine in inhibiting ILC2 function via metabolic restriction, thereby ameliorating key features of asthma pathogenesis.


Assuntos
Asma , Imunidade Inata , Humanos , Dopamina , Linfócitos , Inflamação , Citocinas/metabolismo
5.
J Exp Med ; 218(12)2021 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-34665220

RESUMO

T follicular helper cell (TFH)-dependent antibody responses are critical for long-term immunity. Antibody responses are diminished in early life, limiting long-term protective immunity and allowing prolonged or recurrent infection, which may be important for viral lung infections that are highly prevalent in infancy. In a murine model using respiratory syncytial virus (RSV), we show that TFH and the high-affinity antibody production they promote are vital for preventing disease on RSV reinfection. Following a secondary RSV infection, TFH-deficient mice had significantly exacerbated disease characterized by delayed viral clearance, increased weight loss, and immunopathology. TFH generation in early life was compromised by heightened IL-2 and STAT5 signaling in differentiating naive T cells. Neutralization of IL-2 during early-life RSV infection resulted in a TFH-dependent increase in antibody-mediated immunity and was sufficient to limit disease severity upon reinfection. These data demonstrate the importance of TFH in protection against recurrent RSV infection and highlight a mechanism by which this is suppressed in early life.


Assuntos
Interleucina-2/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Células T Auxiliares Foliculares/imunologia , Células T Auxiliares Foliculares/virologia , Fatores Etários , Animais , Anticorpos Antivirais , Subpopulações de Linfócitos B/imunologia , Subpopulações de Linfócitos B/metabolismo , Subpopulações de Linfócitos B/virologia , Feminino , Centro Germinativo/citologia , Centro Germinativo/imunologia , Centro Germinativo/virologia , Imunidade Humoral , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-2/fisiologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Gravidez , Proteínas Proto-Oncogênicas c-bcl-6/genética , Proteínas Proto-Oncogênicas c-bcl-6/imunologia , Reinfecção/imunologia , Reinfecção/virologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Fator de Transcrição STAT5/metabolismo
6.
Methods Mol Biol ; 2121: 99-114, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32147790

RESUMO

ILC2s are a rare innate cell population capable of rapidly producing type 2 cytokines prior to the recruitment and expansion of adaptive type 2 T helper cells. As a result, they are implicated in the pathogenesis of many type-2 immune-mediated diseases, including allergic airway inflammation. Here we describe methods for interrogating and analyzing ILC2 biology in the context of allergic airway inflammation, such as flow cytometric analysis of mouse and human ILC2s as well as live imaging of pulmonary ILC2s.


Assuntos
Asma/imunologia , Citometria de Fluxo/métodos , Imunidade Inata , Inflamação/imunologia , Microscopia Intravital/métodos , Pulmão/citologia , Subpopulações de Linfócitos/imunologia , Animais , Asma/patologia , Lavagem Broncoalveolar , Citocinas/metabolismo , Humanos , Inflamação/patologia , Microscopia Intravital/instrumentação , Pulmão/diagnóstico por imagem , Pulmão/imunologia , Pulmão/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Subpopulações de Linfócitos/citologia , Camundongos
7.
J Allergy Clin Immunol ; 145(2): 666-678.e9, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31445933

RESUMO

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.


Assuntos
Asma/imunologia , Interferon gama/imunologia , Interleucina-10/imunologia , Células Mieloides/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Alérgenos/imunologia , Animais , Modelos Animais de Doenças , Feminino , Hipersensibilidade/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pyroglyphidae/imunologia
8.
Sci Immunol ; 4(41)2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31704734

RESUMO

Neutrophil mobilization, recruitment, and clearance must be tightly regulated as overexuberant neutrophilic inflammation is implicated in the pathology of chronic diseases, including asthma. Efforts to target neutrophils therapeutically have failed to consider their pleiotropic functions and the implications of disrupting fundamental regulatory pathways that govern their turnover during homeostasis and inflammation. Using the house dust mite (HDM) model of allergic airway disease, we demonstrate that neutrophil depletion unexpectedly resulted in exacerbated T helper 2 (TH2) inflammation, epithelial remodeling, and airway resistance. Mechanistically, this was attributable to a marked increase in systemic granulocyte colony-stimulating factor (G-CSF) concentrations, which are ordinarily negatively regulated in the periphery by transmigrated lung neutrophils. Intriguingly, we found that increased G-CSF augmented allergic sensitization in HDM-exposed animals by directly acting on airway type 2 innate lymphoid cells (ILC2s) to elicit cytokine production. Moreover, increased systemic G-CSF promoted expansion of bone marrow monocyte progenitor populations, which resulted in enhanced antigen presentation by an augmented peripheral monocyte-derived dendritic cell pool. By modeling the effects of neutrophil depletion, our studies have uncovered previously unappreciated roles for G-CSF in modulating ILC2 function and antigen presentation. More broadly, they highlight an unexpected regulatory role for neutrophils in limiting TH2 allergic airway inflammation.


Assuntos
Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Hipersensibilidade/imunologia , Imunidade Inata/imunologia , Inflamação/imunologia , Linfócitos/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Animais , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C
9.
Sci Immunol ; 4(36)2019 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-31175176

RESUMO

Group 2 innate lymphoid cells (ILC2s) are enriched in mucosal tissues (e.g., lung) and respond to epithelial cell-derived cytokines initiating type 2 inflammation. During inflammation, ILC2 numbers are increased in the lung. However, the mechanisms controlling ILC2 trafficking and motility within inflamed lungs remain unclear and are crucial for understanding ILC2 function in pulmonary immunity. Using several approaches, including lung intravital microscopy, we demonstrate that pulmonary ILC2s are highly dynamic, exhibit amoeboid-like movement, and aggregate in the lung peribronchial and perivascular spaces. They express distinct chemokine receptors, including CCR8, and actively home to CCL8 deposits located around the airway epithelium. Within lung tissue, ILC2s were particularly motile in extracellular matrix-enriched regions. We show that collagen-I drives ILC2 to markedly change their morphology by remodeling their actin cytoskeleton to promote environmental exploration critical for regulating eosinophilic inflammation. Our study provides previously unappreciated insights into ILC2 migratory patterns during inflammation and highlights the importance of environmental guidance cues in the lung in controlling ILC2 dynamics.


Assuntos
Pulmão/imunologia , Linfócitos/imunologia , Animais , Movimento Celular/efeitos dos fármacos , Colágeno/imunologia , Eosinófilos/imunologia , Matriz Extracelular/imunologia , Feminino , Fibronectinas/imunologia , Humanos , Imunidade Inata , Inflamação/imunologia , Interleucina-33/farmacologia , Linfócitos/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Proteínas Recombinantes/farmacologia
10.
Eur Respir J ; 54(2)2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31164437

RESUMO

Children with severe therapy-resistant asthma (STRA) have poor control despite maximal treatment, while those with difficult asthma (DA) have poor control from failure to implement basic management, including adherence to therapy. Although recognised as clinically distinct, the airway molecular phenotype, including the role of innate lymphoid cells (ILCs) and their response to steroids in DA and STRA is unknown.Immunophenotyping of sputum and blood ILCs and T-cells from STRA, DA and non-asthmatic controls was undertaken. Leukocytes were analysed longitudinally pre- and post-intramuscular triamcinolone in children with STRA. Cultured ILCs were evaluated to assess steroid responsiveness in vitroAirway eosinophils, type 2 T-helper (Th2) cells and ILC2s were significantly higher in STRA patients compared to DA and disease controls, while IL-17+ lymphoid cells were similar. ILC2s and Th2 cells were significantly reduced in vivo following intramuscular triamcinolone and in vitro with steroids. Furthermore, asthma attacks and symptoms reduced after systemic steroids despite persistence of steroid-resistant IL-17+ cells and eosinophils.Paediatric STRA and DA have distinct airway molecular phenotypes with STRA characterised by elevated type-2 cells. Systemic corticosteroids, but not maintenance inhaled steroids resulted in improved symptom control and exacerbations concomitant with a reduction in functional ILC2s despite persistently elevated IL-17+ lymphoid cells.


Assuntos
Asma/fisiopatologia , Linfócitos/imunologia , Esteroides/uso terapêutico , Células Th2/imunologia , Adolescente , Asma/terapia , Criança , Eosinófilos/imunologia , Feminino , Humanos , Imunidade Inata , Imunofenotipagem , Interleucina-13/metabolismo , Interleucina-17/metabolismo , Leucócitos/imunologia , Leucócitos Mononucleares/imunologia , Pulmão , Masculino , Pediatria , Fenótipo , Células Th17/imunologia , Triancinolona/uso terapêutico
11.
Front Immunol ; 10: 466, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30930901

RESUMO

Vesicular stomatitis virus (VSV) is an insect-transmitted rhabdovirus that is neurovirulent in mice. Upon peripheral VSV infection, CD169+ subcapsular sinus (SCS) macrophages capture VSV in the lymph, support viral replication, and prevent CNS neuroinvasion. To date, the precise mechanisms controlling VSV infection in SCS macrophages remain incompletely understood. Here, we show that Toll-like receptor-7 (TLR7), the main sensing receptor for VSV, is central in controlling lymph-borne VSV infection. Following VSV skin infection, TLR7-/- mice display significantly less VSV titers in the draining lymph nodes (dLN) and viral replication is attenuated in SCS macrophages. In contrast to effects of TLR7 in impeding VSV replication in the dLN, TLR7-/- mice present elevated viral load in the brain and spinal cord highlighting their susceptibility to VSV neuroinvasion. By generating novel TLR7 floxed mice, we interrogate the impact of cell-specific TLR7 function in anti-viral immunity after VSV skin infection. Our data suggests that TLR7 signaling in SCS macrophages supports VSV replication in these cells, increasing LN infection and may account for the delayed onset of VSV-induced neurovirulence observed in TLR7-/- mice. Overall, we identify TLR7 as a novel and essential host factor that critically controls anti-viral immunity to VSV. Furthermore, the novel mouse model generated in our study will be of valuable importance to shed light on cell-intrinsic TLR7 biology in future studies.


Assuntos
Macrófagos/imunologia , Glicoproteínas de Membrana/imunologia , Infecções por Rhabdoviridae/imunologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Receptor 7 Toll-Like/imunologia , Vesiculovirus/fisiologia , Replicação Viral/imunologia , Animais , Encéfalo/imunologia , Encéfalo/virologia , Macrófagos/virologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Infecções por Rhabdoviridae/genética , Infecções por Rhabdoviridae/patologia , Lectina 1 Semelhante a Ig de Ligação ao Ácido Siálico/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Medula Espinal/imunologia , Medula Espinal/virologia , Receptor 7 Toll-Like/genética , Replicação Viral/genética
12.
Immunol Cell Biol ; 97(3): 246-257, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30768869

RESUMO

The lungs present a challenging immunological dilemma for the host. Anatomically positioned at the environmental interface, they are constantly exposed to antigens, pollutants and microbes, while simultaneously facilitating vital gas exchange. Remarkably, the lungs maintain a functionally healthy state, ignoring harmless inhaled proteins, adapting to toxic environmental insults and limiting immune responses to allergens and pathogenic microbes. This functional strategy of environmental adaptation maintains immune defense, reduces tissue damage, and promotes and sustains lung immune tolerance. At steady state, airway macrophages produce low levels of cytokines, and suppress the induction of innate and adaptive immunity. These cells are primary initiators of lung innate immunity and possess high phagocytic activity to clear particulate antigens and apoptotic cell debris from the airways to regulate the response to infection and inflammation. In response to epithelial injury, resident and recruited macrophages drive tissue repair. In this review, we will focus on the functional importance of macrophages in tissue homeostasis and inflammation in the lung and highlight how environmental cues alter the plasticity and function of lung airway macrophages. We will also discuss mechanisms employed by pulmonary macrophages to promote resolution of tissue inflammation, and how and when this balance is perturbed, they contribute to pathological remodeling in acute and chronic infections and diseases such as asthma, idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease.


Assuntos
Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Cicatrização , Remodelação das Vias Aéreas/imunologia , Animais , Biomarcadores , Microambiente Celular/imunologia , Suscetibilidade a Doenças , Homeostase , Interações Hospedeiro-Patógeno/imunologia , Humanos , Pulmão/patologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Especificidade de Órgãos/imunologia , Fenótipo , Pneumonia/etiologia , Pneumonia/metabolismo , Pneumonia/patologia , Regeneração
13.
Front Immunol ; 10: 3114, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32038635

RESUMO

Group 2 innate lymphoid cells (ILC2s) are enriched at mucosal sites, including the lung, and play a central role in type 2 immunity and maintaining tissue homeostasis. As a result, since their discovery in 2010, research into ILC2s has increased markedly. Numerous strategies have been used to define ILC2s by flow cytometry, often utilizing different combinations of surface markers despite their expression being variable and context-dependent. In this study, we sought to generate a comprehensive characterization of pulmonary ILC2s, identifying stable and context specific markers from different pulmonary compartments following different airway exposures in different strains of mice. Our analysis revealed that pulmonary ILC2 surface marker phenotype is heterogeneous and is influenced by mouse strain, pulmonary location, in vivo treatment/exposure and ex vivo stimulation. Therefore, we propose that a lineage negative cell expressing CD45 and Gata3 defines an ILC2, and subsequent surface marker expression should be used to describe their phenotype in context-specific scenarios.


Assuntos
Biomarcadores , Imunidade Inata , Imunofenotipagem , Linfócitos/imunologia , Linfócitos/metabolismo , Alérgenos/imunologia , Animais , Feminino , Imuno-Histoquímica , Interleucina-33/metabolismo , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Especificidade da Espécie
14.
Cell Rep ; 17(4): 1113-1127, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760315

RESUMO

Cytomegalovirus (CMV) is an opportunistic virus severely infecting immunocompromised individuals. In mice, endosomal Toll-like receptor 9 (TLR9) and downstream myeloid differentiation factor 88 (MyD88) are central to activating innate immune responses against mouse CMV (MCMV). In this respect, the cell-specific contribution of these pathways in initiating anti-MCMV immunity remains unclear. Using transgenic mice, we demonstrate that TLR9/MyD88 signaling selectively in CD11c+ dendritic cells (DCs) strongly enhances MCMV clearance by boosting natural killer (NK) cell CD69 expression and IFN-γ production. In addition, we show that in the absence of plasmacytoid DCs (pDCs), conventional DCs (cDCs) promote robust NK cell effector function and MCMV clearance in a TLR9/MyD88-dependent manner. Simultaneously, cDC-derived IL-15 regulates NK cell degranulation by TLR9/MyD88-independent mechanisms. Overall, we compartmentalize the cellular contribution of TLR9 and MyD88 signaling in individual DC subsets and evaluate the mechanism by which cDCs control MCMV immunity.


Assuntos
Infecções por Citomegalovirus/prevenção & controle , Infecções por Citomegalovirus/virologia , Células Dendríticas/metabolismo , Muromegalovirus/fisiologia , Fator 88 de Diferenciação Mieloide/metabolismo , Transdução de Sinais , Receptor Toll-Like 9/metabolismo , Animais , Antivirais/farmacologia , Antígeno CD11c/metabolismo , Citotoxicidade Imunológica , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos BALB C
15.
Oncoimmunology ; 4(8): e970462, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26405564

RESUMO

Therapeutic vaccinations against cancer are still largely ineffective. Major caveats are inefficient delivery of tumor antigens to dendritic cells (DCs) and excessive immune suppression by Foxp3+ regulatory T cells (Tregs), resulting in defective T cell priming and failure to induce tumor regression. To circumvent these problems we evaluated a novel combinatorial therapeutic strategy. We show that tumor antigen targeting to DC-SIGN in humanized hSIGN mice via glycans or specific antibodies induces superior T cell priming. Next, this targeted therapy was combined with transient Foxp3+ Treg depletion employing hSIGNxDEREG mice. While Treg depletion alone slightly delayed B16-OVA melanoma growth, only the combination therapy instigated long-term tumor regression in a substantial fraction of mice. This novel strategy resulted in optimal generation of antigen-specific activated CD8+ T cells which accumulated in regressing tumors. Notably, Treg depletion also allowed the local appearance of effector T cells specific for endogenous B16 antigens. This indicates that antitumor immune responses can be broadened by therapies aimed at controlling Tregs in tumor environments. Thus, transient inhibition of Treg-mediated immune suppression potentiates DC targeted antigen vaccination and tumor-specific immunity.

16.
Blood ; 124(20): 3081-91, 2014 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-25100743

RESUMO

Multiple subsets of FMS-like tyrosine kinase 3 ligand (FLT3L)-dependent dendritic cells (DCs) control T-cell tolerance and immunity. In mice, Batf3-dependent CD103(+) DCs efficiently enter lymph nodes and cross-present antigens, rendering this conserved DC subset a promising target for tolerance induction or vaccination. However, only limited numbers of CD103(+) DCs can be isolated with current methods. Established bone marrow culture protocols efficiently generate monocyte-derived DCs or produce a mixture of FLT3L-dependent DC subsets. We show that CD103(+) DC development requires prolonged culture time and continuous action of both FLT3L and granulocyte macrophage colony-stimulating factor (GM-CSF), explained by a dual effect of GM-CSF on DC precursors and differentiating CD103(+) DCs. Accordingly, we established a novel method to generate large numbers of CD103(+) DCs (iCD103-DCs) with limited presence of other DC subsets. iCD103-DCs develop in a Batf3- and Irf8-dependent fashion, express a CD8α/CD103 DC gene signature, cross-present cell-associated antigens, and respond to TLR3 stimulation. Thus, iCD103-DCs reflect key features of tissue CD103(+) DCs. Importantly, iCD103-DCs express high levels of CCR7 upon maturation and migrate to lymph nodes more efficiently than classical monocyte-derived DCs. Finally, iCD103-DCs induce T cell-mediated protective immunity in vivo. Our study provides insights into CD103(+) DC development and function.


Assuntos
Antígenos CD/imunologia , Fatores de Transcrição de Zíper de Leucina Básica/imunologia , Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Células Dendríticas/citologia , Células Dendríticas/imunologia , Cadeias alfa de Integrinas/imunologia , Proteínas Repressoras/imunologia , Animais , Antígenos CD/análise , Fatores de Transcrição de Zíper de Leucina Básica/análise , Diferenciação Celular , Células Cultivadas , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Imunidade Celular , Cadeias alfa de Integrinas/análise , Proteínas de Membrana/imunologia , Camundongos , Proteínas Repressoras/análise , Linfócitos T/imunologia , Receptor 3 Toll-Like/imunologia
17.
Virol J ; 11: 145, 2014 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-25108672

RESUMO

BACKGROUND: Cytomegalovirus establishes lifelong persistency in the host and leads to life threatening situations in immunocompromised patients. FoxP3+ T regulatory cells (Tregs) critically control and suppress innate and adaptive immune responses. However, their specific role during MCMV infection, especially pertaining to their interaction with NK cells, remains incompletely defined. METHODS: To understand the contribution of Tregs on NK cell function during acute MCMV infection, we infected Treg depleted and undepleted DEREG mice with WT MCMV and examined Treg and NK cell frequency, number, activation and effector function in vivo. RESULTS: Our results reveal an increased frequency of activated Tregs within the CD4+ T cell population shortly after MCMV infection. Specific depletion of Tregs in DEREG mice under homeostatic conditions leads to an increase in NK cell number as well as to a higher activation status of these cells as compared with non-depleted controls. Interestingly, upon infection this effect on NK cells is completely neutralized in terms of cell frequency, CD69 expression and functionality with respect to IFN-γ production. Furthermore, composition of the NK cell population with regard to Ly49H expression remains unchanged. In contrast, absence of Tregs still boosts the general T cell response upon infection to a level comparable to the enhanced activation seen in uninfected mice. CD4+ T cells especially benefit from Treg depletion exhibiting a two-fold increase of CD69+ cells 40 h and IFN-γ+ cells 7 days p.i. while, MCMV infection per se induces robust CD8+ T cell activation which is also further augmented in Treg-depleted mice. Nevertheless, the viral burden in the liver and spleen remain unaltered upon Treg ablation during the course of infection. CONCLUSIONS: Thus, MCMV infection abolishes Treg suppressing effects on NK cells whereas T cells benefit from their absence during acute infection. This study provides novel information in understanding the collaborative interaction between NK cells and Tregs during a viral infection and provides further knowledge that could be adopted in therapeutic setups to improve current treatment of organ transplant patients where modulation of Tregs is envisioned as a strategy to overcome transplant rejection.


Assuntos
Infecções por Herpesviridae/imunologia , Células Matadoras Naturais/imunologia , Muromegalovirus/imunologia , Linfócitos T Reguladores/imunologia , Imunidade Adaptativa , Animais , Fatores de Transcrição Forkhead/metabolismo , Infecções por Herpesviridae/virologia , Homeostase/imunologia , Interferon gama/biossíntese , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Depleção Linfocítica , Masculino , Camundongos , Fenótipo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Carga Viral
18.
PLoS Pathog ; 9(9): e1003648, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086137

RESUMO

Plasmacytoid dendritic cells (pDCs) express the I-type lectin receptor Siglec-H and produce interferon α (IFNα), a critical anti-viral cytokine during the acute phase of murine cytomegalovirus (MCMV) infection. The ligands and biological functions of Siglec-H still remain incompletely defined in vivo. Thus, we generated a novel bacterial artificial chromosome (BAC)-transgenic "pDCre" mouse which expresses Cre recombinase under the control of the Siglec-H promoter. By crossing these mice with a Rosa26 reporter strain, a representative fraction of Siglec-H⁺ pDCs is terminally labeled with red fluorescent protein (RFP). Interestingly, systemic MCMV infection of these mice causes the downregulation of Siglec-H surface expression. This decline occurs in a TLR9- and MyD88-dependent manner. To elucidate the functional role of Siglec-H during MCMV infection, we utilized a novel Siglec-H deficient mouse strain. In the absence of Siglec-H, the low infection rate of pDCs with MCMV remained unchanged, and pDC activation was still intact. Strikingly, Siglec-H deficiency induced a significant increase in serum IFNα levels following systemic MCMV infection. Although Siglec-H modulates anti-viral IFNα production, the control of viral replication was unchanged in vivo. The novel mouse models will be valuable to shed further light on pDC biology in future studies.


Assuntos
Células Dendríticas/imunologia , Infecções por Herpesviridae/imunologia , Interferon-alfa/imunologia , Lectinas/imunologia , Modelos Imunológicos , Muromegalovirus/fisiologia , Plasmócitos/imunologia , Receptores de Superfície Celular/imunologia , Animais , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/patologia , Interferon-alfa/genética , Interferon-alfa/metabolismo , Lectinas/genética , Lectinas/metabolismo , Camundongos , Camundongos Knockout , Plasmócitos/metabolismo , Plasmócitos/patologia , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Replicação Viral/genética , Replicação Viral/imunologia
19.
PLoS One ; 7(10): e47102, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23071726

RESUMO

Regulatory T cells (Tregs) play a non-redundant role in maintenance of immune homeostasis. This is achieved by suppressing both, priming of naïve cells and effector cell functions. Although Tregs have been implicated in modulating allergic immune responses, their influence on distinct phases of development of allergies remains unclear. In this study, by using bacterial artificial chromosome (BAC)-transgenic Foxp3-DTR (DEREG) mice we demonstrate that the absence of Foxp3(+) Tregs during the allergen challenge surprisingly does not exacerbate allergic airway inflammation in BALB/c mice. As genetic disposition due to strain specificity may contribute significantly to development of allergies, we performed similar experiment in C57BL/6 mice, which are less susceptible to allergy in the model of sensitization used in this study. We report that the genetic background does not influence the consequence of this depletion regimen. These results signify the temporal regulation exerted by Foxp3(+) Tregs in limiting allergic airway inflammation and may influence their application as potential therapeutics.


Assuntos
Fatores de Transcrição Forkhead/genética , Inflamação/imunologia , Hipersensibilidade Respiratória/imunologia , Linfócitos T Reguladores/imunologia , Animais , Cromossomos Artificiais Bacterianos , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Fatores de Transcrição Forkhead/imunologia , Fatores de Transcrição Forkhead/metabolismo , Predisposição Genética para Doença , Inflamação/genética , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/imunologia , Ovalbumina/toxicidade , Linfócitos T Reguladores/metabolismo
20.
Microb Biotechnol ; 5(2): 260-9, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21951341

RESUMO

The development of effective vaccines against life-threatening pathogens in human diseases represents one of the biggest challenges in biomedical science. Vaccines traditionally make use of the body's own immune armoury to combat pathogens. Yet, while our immune system is mostly effective in eliminating or controlling a diverse range of microorganisms, its responses are incomplete or somewhat limited in several other cases. How immune responses are restrained during certain infections has been a matter of debate for many years. The discovery of regulatory T cells (Tregs), an immune cell type that plays a central role in maintaining immune homeostasis and controlling appropriate immune responses, has shed light into many questions. Indeed, it has been proposed that while Tregs might be beneficial in preventing excessive tissue damage during infection, they might also favour pathogen persistence by restraining effector immune responses. In addition, Tregs are believed to limit immune responses upon vaccination. Different strategies have been pursued to circumvent Treg activity during immunization, but the lack of specific tools for their study has led sometimes to controversial conclusions. With the advent of novel mouse models that allow specific depletion and/or tracking of Treg populations in vivo, novel aspects of Treg biology during infection have been unravelled. In this review, we describe the new advances in understanding Treg biology during infection and evaluate Treg depletion as a novel adjuvant strategy for vaccination.


Assuntos
Tolerância Imunológica , Infecções/imunologia , Linfócitos T Reguladores/imunologia , Vacinas/imunologia , Animais , Homeostase , Humanos , Infecções/patologia , Depleção Linfocítica , Camundongos , Modelos Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...