Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 180
Filter
Add more filters










Publication year range
1.
Mucosal Immunol ; 10(6): 1569-1580, 2017 11.
Article in English | MEDLINE | ID: mdl-28295020

ABSTRACT

Regulation of respiratory mucosal immunity by microbial-derived metabolites has been a proposed mechanism that may provide airway protection. Here we examine the effect of oral Lactobacillus johnsonii supplementation on metabolic and immune response dynamics during respiratory syncytial virus (RSV) infection. L. johnsonii supplementation reduced airway T helper type 2 cytokines and dendritic cell (DC) function, increased regulatory T cells, and was associated with a reprogrammed circulating metabolic environment, including docosahexanoic acid (DHA) enrichment. RSV-infected bone marrow-derived DCs (BMDCs) from L. johnsonii-supplemented mice had altered cytokine secretion, reduced expression of co-stimulatory molecules, and modified CD4+ T-cell cytokines. This was replicated upon co-incubation of wild-type BMDCs with either plasma from L. johnsonii-supplemented mice or DHA. Finally, airway transfer of BMDCs from L. johnsonii-supplemented mice or with wild-type derived BMDCs pretreated with plasma from L. johnsonii-supplemented mice reduced airway pathological responses to infection in recipient animals. Thus L. johnsonii supplementation mediates airway mucosal protection via immunomodulatory metabolites and altered immune function.


Subject(s)
Bone Marrow Cells/immunology , Dendritic Cells/immunology , Lactobacillus johnsonii/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/metabolism , Animals , Bone Marrow Cells/virology , Cell Line , Cellular Microenvironment , Cellular Reprogramming , Cytokines/metabolism , Dendritic Cells/virology , Dietary Supplements , Docosahexaenoic Acids/metabolism , Immunomodulation , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections/prevention & control , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology
2.
Mucosal Immunol ; 10(2): 299-306, 2017 03.
Article in English | MEDLINE | ID: mdl-27966551

ABSTRACT

The lungs are not sterile or free from bacteria; rather, they harbor a distinct microbiome whose composition is driven by different ecological rules than for the gastrointestinal tract. During disease, there is often a shift in community composition towards Gammaproteobacteria, the bacterial class that contains many common lung-associated gram-negative "pathogens." Numerous byproducts of host inflammation are growth factors for these bacteria. The extracellular nutrient supply for bacteria in the lungs, which is severely limited during health, markedly increases due to the presence of mucus and vascular permeability. While Gammaproteobacteria benefit from airway inflammation, they also encode molecular components that promote inflammation, potentially creating a cyclical inflammatory mechanism. In contrast, Prevotella species that are routinely acquired via microaspiration from the oral cavity may participate in immunologic homeostasis of the airways.vAreas of future research include determining for specific lung diseases (1) whether an altered lung microbiome initiates disease pathogenesis, promotes chronic inflammation, or is merely a marker of injury and inflammation, (2) whether the lung microbiome can be manipulated therapeutically to change disease progression, (3) what molecules (metabolites) generated during an inflammatory response promote cross-kingdom signaling, and (4) how the lung "ecosystem" collapses during pneumonia, to be dominated by a single pathogen.


Subject(s)
Dysbiosis/immunology , Gammaproteobacteria/immunology , Lung Diseases/microbiology , Microbiota , Pneumonia/microbiology , Prevotella/immunology , Respiratory System/microbiology , Animals , Capillary Permeability , Homeostasis , Host-Pathogen Interactions , Humans , Lung Diseases/immunology , Pneumonia/immunology , Respiratory System/immunology
3.
Mucosal Immunol ; 10(3): 673-684, 2017 05.
Article in English | MEDLINE | ID: mdl-27624780

ABSTRACT

Specific components of the intestinal microbiota are capable of influencing immune responses such that a mutualistic relationship is established. In mice, colonization with segmented filamentous bacteria (SFB) induces T-helper-17 (Th17) cell differentiation in the intestine, yet the effector functions of interleukin (IL)-17A in response to SFB remain incompletely understood. Here we report that colonization of mice with SFB-containing microbiota induced IL-17A- and CXCR2-dependent recruitment of neutrophils to the ileum. This response required adaptive immunity, as Rag-deficient mice colonized with SFB-containing microbiota failed to induce IL-17A, CXCL1 and CXCL2, and displayed defective neutrophil recruitment to the ileum. Interestingly, neutrophil depletion in wild-type mice resulted in significantly augmented Th17 responses and SFB expansion, which correlated with impaired expression of IL-22 and antimicrobial peptides. These data provide novel insight into a dynamic IL-17A-CXCR2-neutrophil axis during acute SFB colonization and demonstrate a central role for neutrophils in limiting SFB expansion.


Subject(s)
Bacteria/immunology , Gastrointestinal Microbiome/immunology , Ileum/immunology , Interleukin-17/metabolism , Neutrophils/immunology , Receptors, Interleukin-8B/metabolism , Th17 Cells/immunology , Adaptive Immunity/genetics , Animals , Antimicrobial Cationic Peptides/metabolism , Bacteria/growth & development , Cell Differentiation , Cell Movement/genetics , Cells, Cultured , Homeodomain Proteins/genetics , Ileum/microbiology , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Interleukin-22
4.
Mucosal Immunol ; 9(3): 610-20, 2016 05.
Article in English | MEDLINE | ID: mdl-26376362

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) efficacy is limited by numerous pulmonary complications. We developed a model of syngeneic bone marrow transplantion (BMT) followed by infection with murine gamma herpesvirus-68 that results in pneumonitis and fibrosis and mimics human "noninfectious" HSCT complications. BMT mice experience increased early lytic replication, but establish viral latency by 21 days post infection. CD4 T cells in BMT mice are skewed toward interleukin (IL)-17A rather than interferon (IFN)-γ production. Transplantation of bone marrow from Il-17a(-/-) donors or treatment with anti-IL-17A neutralization antibodies at late stages attenuates pneumonitis and fibrosis in infected BMT mice, suggesting that hematopoietic-derived IL-17A is essential for development of pathology. IL-17A directly influences activation and extracellular matrix production by lung mesenchymal cells. Lung CD11c+ cells of BMT mice secrete more transforming growth factor beta-ß1, and pro-TH17 mRNAs for IL-23 and IL-6, and less TH1-promoting cytokine mRNA for IFN-γ but slightly more IL-12 mRNA in response to viral infection. Adoptive transfer of non-BMT lung CD11c-enriched cells restores robust TH1 response and suppresses aberrant TH17 response in BMT mice to improve lung pathology. Our data suggest that "noninfectious" HSCT lung complications may reflect preceding viral infections and demonstrate that IL-17A neutralization may offer therapeutic advantage even after disease onset.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Transplantation , Herpesviridae Infections/immunology , Lung/pathology , Pneumonia/immunology , Postoperative Complications/immunology , Rhadinovirus/physiology , Th17 Cells/immunology , Animals , Antibodies, Neutralizing/administration & dosage , Cells, Cultured , Disease Models, Animal , Fibrosis , Humans , Interleukin-17/genetics , Interleukin-17/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/etiology , Pneumonia/prevention & control , Postoperative Complications/prevention & control , Th17 Cells/virology , Virus Latency , Virus Replication
5.
Mucosal Immunol ; 8(5): 1118-30, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25669150

ABSTRACT

While recent studies suggest that interleukin (IL)-1ß production is modulated by macroautophagy or sensors of endoplasmic reticulum (ER) stress upon pro-inflammatory insult, autophagy and IL-1ß production during viral infection has not been fully investigated. This was addressed using respiratory syncytial virus (RSV), which is associated with lung immunopathology, IL-1, and IL-17a secretion in severely infected patients. Mice deficient in the autophagy-associated protein Map1-LC3b (LC3b(-/-)) developed increased IL-17a-dependent lung pathology upon infection. RSV-infected LC3b(-/-) dendritic cells (DCs) fail to upregulate autophagosome formation, secrete IL-1ß and IL-6, and elicit IL-17a production from CD4+ T cells. Bone marrow chimeras revealed that both structural and hematopoietic LC3b deficiency contribute to the development of IL-17a-dependent lung pathology in vivo. Further investigation revealed airway epithelium as the primary source of IL-1ß during infection, whereas inhibition of the ER-stress sensor inositol-requiring protein-1 in primary airway epithelial cells reduced IL-1ß production identifying a primary ER stress pathway. Finally, blockade of IL-1 receptor signaling in RSV-infected LC3b(-/-) mice abolished IL-17a-dependent lung pathology. These findings provide novel mechanistic insight into the contribution of autophagy- and ER stress-dependent cytokine production that initiate and maintain aberrant Th17 responses, while identifying IL-1 as a potential therapeutic target in the treatment of severe respiratory viral infections.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Interleukin-17/immunology , Interleukin-1beta/immunology , Lung Diseases/immunology , Microtubule-Associated Proteins/deficiency , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/immunology , Endoplasmic Reticulum Stress/genetics , Interleukin-17/genetics , Interleukin-1beta/genetics , Interleukin-6/genetics , Interleukin-6/immunology , Lung Diseases/genetics , Lung Diseases/pathology , Mice , Mice, Knockout , Microtubule-Associated Proteins/immunology , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/pathology , Th17 Cells/immunology , Th17 Cells/pathology
6.
Mucosal Immunol ; 8(5): 1131-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25669152

ABSTRACT

The generation of regulatory T (Treg) cells is driven by Foxp3 and is responsible for dampening inflammation and reducing autoimmunity. In this study, the epigenetic regulation of inducible Treg (iTreg) cells was examined and an H3K4 histone methyltransferase, SMYD3 (SET and MYND Domain 3), which regulates the expression of Foxp3 by a TGFß1/Smad3 (transforming growth factor-ß1/Smad3)-dependent mechanism, was identified. Using chromatin immunoprecipitation assays, SMYD3 depletion led to a reduction in H3K4me3 in the promoter region and CNS1 (conserved noncoding DNA sequence) of the foxp3 locus. SMYD3 abrogation affected iTreg cell formation while allowing dysregulated interleukin-17 production. In a mouse model of respiratory syncytial virus (RSV) infection, a model in which iTreg cells have a critical role in regulating lung pathogenesis, SMYD3(-/-) mice demonstrated exacerbation of RSV-induced disease related to enhanced proinflammatory responses and worsened pathogenesis within the lung. Our data highlight a novel activation role for the TGFß-inducible SMYD3 in regulating iTreg cell formation leading to increased severity of virus-related disease.


Subject(s)
Epigenesis, Genetic/immunology , Forkhead Transcription Factors/immunology , Histone-Lysine N-Methyltransferase/immunology , Lung Diseases/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Forkhead Transcription Factors/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Lung Diseases/genetics , Lung Diseases/pathology , Mice , Mice, Knockout , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/pathology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology
7.
Int Immunopharmacol ; 22(1): 242-7, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24998164

ABSTRACT

Upper airway viral infection in patients with airway allergy often exacerbates olfactory dysfunction, but the mechanism for this exacerbation remains unclear. Here, we examined the effects of respiratory syncytial virus (RSV) infection, in the presence or absence of airway allergy, on olfactory receptor neurons (ORNs) and their progenitors in mice. Immunohistological analyses revealed that cockroach allergen (CRA)-induced airway allergy alone did not affect the number of OMP(+) mature ORNs and SOX2(+) ORN progenitors. Intranasal RSV line 19 infection in allergy-free mice resulted in a transient decrease in SOX2(+) ORN progenitors without affecting OMP(+) ORNs. In contrast, the RSV-induced decrease in SOX2(+) ORN progenitors was exacerbated and prolonged in allergic mice, which resulted in eventual loss of OMP(+) ORNs. In the allergic mice, reduction of RSV in the olfactory epithelium was delayed as compared with allergy-free mice. These results suggest that ORN progenitors were impaired by RSV infection and that airway allergy exacerbated damage to ORN progenitors by reducing viral clearance.


Subject(s)
Hypersensitivity/immunology , Nasal Mucosa/immunology , Olfactory Receptor Neurons/physiology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Allergens/immunology , Animals , Cell Differentiation/immunology , Cockroaches , Female , Hypersensitivity/complications , Mice , Mice, Inbred BALB C , Nasal Mucosa/virology , Olfactory Receptor Neurons/virology , Respiratory Syncytial Virus Infections/complications , SOXB1 Transcription Factors/metabolism , Viral Load
8.
Clin Exp Immunol ; 172(2): 263-79, 2013 May.
Article in English | MEDLINE | ID: mdl-23574323

ABSTRACT

RSV lower respiratory tract infections (LRTI) are among the most common diseases necessitating hospital admission in children. In addition to causing acute respiratory failure, RSV infections are associated with sequelae such as secondary bacterial infections and reactive airway disease. One characteristic host response observed in severe RSV-induced LRTI and/or subsequent development of asthma is increased expression of interleukin (IL)-10. However, contradictory results have been reported regarding whether IL-10 inhibits asthmatic responses or intensifies the disease. We aimed to reconcile these discordant observations by elucidating the role of IL-10 in regulating the host response to RSV LRTI. In this study, we used a lung-specific, inducible IL-10 over-expression (OE) transgenic mouse model to address this question. Our results showed that the presence of IL-10 at the time of RSV infection not only attenuated acute inflammatory process (i.e. 24 h post-infection), but also late inflammatory changes [characterized by T helper type 2 (Th2) cytokine and chemokine expression]. While this result appears contradictory to some clinical observations where elevated IL-10 levels are observed in asthmatic patients, we also found that delaying IL-10 OE until the late immune response to RSV infection, additive effects rather than inhibitory effects were observed. Importantly, in non-infected, IL-10 OE mice, IL-10 OE alone induced up-regulation of Th2 cytokine (IL-13 and IL-5) and Th2-related chemokine [monocyte chemoattractant protein 1 (MCP-1), chemokine (C-C motif) ligand 3 (CCL3) and regulated upon activation normal T cell expressed and secreted (RANTES)] expression. We identified a subset of CD11b(+)CD11c(+)CD49b(+)F4/80(-)Gr-1(-) myeloid cells as a prinicipal source of IL-10-induced IL-13 production. Therefore, the augmented pathological responses observed in our 'delayed' IL-10 over-expression model could be attributed to IL-10 OE alone. Taken together, our study indicated dual roles of IL-10 on RSV-induced lung inflammation which appear to depend upon the timing of when elevated IL-10 is expressed in the lung.


Subject(s)
Interleukin-10/metabolism , Pneumonia/immunology , Pneumonia/virology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/virology , Chemokine CCL2/genetics , Chemokine CCL5/metabolism , Interleukin-13 , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/metabolism , Respiratory Tract Infections/immunology , Respiratory Tract Infections/virology , Th2 Cells/immunology
9.
Arch Virol ; 150(12): 2501-12, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16012783

ABSTRACT

Barley mild mosaic virus (BaMMV), a member of the family Potyviridae, genus Bymovirus, is involved in the economically important yellow mosaic disease of winter barley in East Asia and Europe. We investigated serological properties of bacterially expressed BaMMV coat protein (CP) of a German isolate. Ten mouse monoclonal antibodies were produced using purified E. coli expressed BaMMV-CP as immunogen. The reactivity of MAbs with different strains of BaMMV was analysed by several immunological methods that are frequently used in diagnostic virology: enzyme-linked immunosorbent assay (ELISA), dot-blot, Western-blotting (WB), direct tissue blotting immunoassay (DTBIA) and immunoelectron microscopy (IEM). The amino acids involved in the formation of epitopes recognised by several MAbs were mapped by using synthetic pin-bound peptides and the localisation of epitopes in assembled virus particles was determined by electron microscope studies. MAbs V29 and M1 decorated the whole virion indicating that their epitopes 6PDPI9 and 96ITDDEK101, respectively, are exposed on the surface. The MAbs V6 and V14 both interacted with 44LPEPKM49, which seems to be accessible at only one end of the virus particle. The MAbs V6, V14, V29 and M1 detected epitopes common to a wide range of BaMMV isolates and can therefore be used effectively in routine diagnostic tests for BaMMV from barley leaves. We suggest that MAbs M1, V6, V14 and V29 are most suitable for use in TAS-ELISA, V6, V14 and V29 for Western blotting and V29 and M1 for electron microscope serology.


Subject(s)
Capsid Proteins/immunology , Epitopes/immunology , Potyviridae/immunology , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes/chemistry , Hordeum/virology , Immunoblotting , Microscopy, Immunoelectron , Plant Diseases/virology
10.
Inflamm Res ; 54(12): 514-21, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16389573

ABSTRACT

OBJECTIVE: To investigate the effect of the X-linked immunodeficiency (Xid) B cell defect on the response to the cockroach allergen in mice. METHODS: Two cockroach allergen immunization and challenge protocols were employed to sensitize CBA/J wild-type and CBA/CaHN-btk(-/-)xid/J (Xid) mice. Blood and tissue samples were collected 24 and 48 hrs after the last intratracheal antigen challenge and were analyzed for several parameters of allergic inflammation. RESULTS: Nearly equivalent amounts of serum IgE were detected in Xid and CBA/J mice after short-term antigen challenge despite the B cell deficiency in Xid mice. A decreased concentration of IgE was detected in CBA/J mice after repeated allergen challenges but not in the Xid mice. Correlating with the discrepancy in serum IgE levels, higher levels of IL-13, IL-5, IL-10 and CCL5 were measured in whole lung homogenates from allergen-challenged Xid mice compared to CBA/J mice. In addition, draining lymph node cells from Xid mice expressed elevated levels of IL-4, IL-5, IL-10 and IFNgamma mRNA compared to cells from CBA/J mice after in vitro culture with cockroach antigen. An increase in lung inflammation, interstitial eosinophilia and mucus production was also observed in allergen-challenged Xid mice. CD95L expression increased on B-1a cells following allergen challenge, which was accompanied by an increase in lung CD4(+) Th cell apoptosis in wild-type CBA/J mice. In contrast, Xid mice did not have an increase in CD4(+) T cell apoptosis following allergen challenge. CONCLUSIONS: These data suggest a regulatory role for B-1a cells in reducing cytokine production, pulmonary inflammation, and CD4(+) T cell survival during cockroach allergen-induced airway inflammation.


Subject(s)
Asthma/immunology , B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/immunology , Hypersensitivity/immunology , Animals , Apoptosis , Asthma/pathology , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD4 Antigens/analysis , CD4 Antigens/physiology , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/physiology , CD5 Antigens/analysis , Chemokine CCL5/metabolism , Cockroaches/immunology , Cytokines/analysis , Disease Models, Animal , Fas Ligand Protein , Female , Gene Expression Regulation , Hypersensitivity/pathology , Immunoglobulin E/blood , Lung/chemistry , Lung/immunology , Lung/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred CBA , Mice, Mutant Strains , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/metabolism
11.
Clin Exp Immunol ; 136(1): 15-20, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15030509

ABSTRACT

Stem cell factor (SCF) has a significant role in the inflammation and activation of allergic airway responses. When monoclonal anti-SCF was administered intratracheally during allergen challenge there was a significant alteration of eosinophil accumulation and airway hyperreactivity (AHR). Anti-SCF treatment also attenuated pulmonary cytokine and chemokine levels. In particular, there was an antibody dose-dependent decrease in interleukin (IL)-5 and tumour necrosis factor (TNF)-alpha. There was also a significant reduction of CCL2 and CCL5, which correlated with the reduction in AHR. Mice treated with anti-SCF demonstrated a significant decrease in pulmonary gob-5 gene expression, which has been shown to correlate to goblet cell hyperplasia/metaplasia relating to airway mucus production. Blocking SCF-mediated activation within the airway using a monoclonal antibody indicates that this cytokine may represent a viable target for therapeutic intervention that could affect multiple aspects of allergen-induced immunopathology.


Subject(s)
Asthma/prevention & control , Cytokines/metabolism , Lung/immunology , Stem Cell Factor/antagonists & inhibitors , Allergens/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Asthma/immunology , Asthma/pathology , Chemokines/metabolism , Chloride Channels/metabolism , Eosinophilia/prevention & control , Methacholine Chloride , Mice , Mice, Inbred CBA , Mucoproteins/metabolism , Stem Cell Factor/immunology
12.
Braz. j. med. biol. res ; 36(11): 1455-1463, Nov. 2003.
Article in English | LILACS | ID: lil-348292

ABSTRACT

Chemokines are important chemotactic cytokines that play a fundamental role in the trafficking of leukocytes to sites of inflammation. They are also potent cell-activating factors, inducing cytokine and histamine release and free radical production, a fact that makes them particularly important in the pathogenesis of allergic inflammation. The action of chemokines is regulated at the level of agonist production and processing as well as at the level of receptor expression and coupling. Therefore, an analysis of the ligands must necessarily consider receptors. Eosinophils are target cells involved in the allergic inflammatory response since they are able to release a wide variety of mediators including CC and CXC chemokines and express their receptors. These mediators could damage the airway epithelial cells and might be important to stimulate other cells inducing an amplification of the allergic response. This review focuses on recently emerging data pertaining to the importance of chemokines and chemokine receptors in promoting eosinophil activation and migration during the allergic inflammatory process. The analysis of the function of eosinophils and their chemokine receptors during allergic inflammation might be a good approach to understanding the determinants of asthma severity and to developing novel therapies.


Subject(s)
Animals , Humans , Asthma , Chemokines , Eosinophils , Receptors, Chemokine , Cell Degranulation , Chemokines , Eosinophils , Receptors, Chemokine , Severity of Illness Index
13.
Braz J Med Biol Res ; 36(11): 1455-63, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14576899

ABSTRACT

Chemokines are important chemotactic cytokines that play a fundamental role in the trafficking of leukocytes to sites of inflammation. They are also potent cell-activating factors, inducing cytokine and histamine release and free radical production, a fact that makes them particularly important in the pathogenesis of allergic inflammation. The action of chemokines is regulated at the level of agonist production and processing as well as at the level of receptor expression and coupling. Therefore, an analysis of the ligands must necessarily consider receptors. Eosinophils are target cells involved in the allergic inflammatory response since they are able to release a wide variety of mediators including CC and CXC chemokines and express their receptors. These mediators could damage the airway epithelial cells and might be important to stimulate other cells inducing an amplification of the allergic response. This review focuses on recently emerging data pertaining to the importance of chemokines and chemokine receptors in promoting eosinophil activation and migration during the allergic inflammatory process. The analysis of the function of eosinophils and their chemokine receptors during allergic inflammation might be a good approach to understanding the determinants of asthma severity and to developing novel therapies.


Subject(s)
Asthma/metabolism , Chemokines/physiology , Eosinophils/physiology , Receptors, Chemokine/physiology , Animals , Cell Degranulation , Chemokines/metabolism , Eosinophils/metabolism , Humans , Hypersensitivity/metabolism , Inflammation/metabolism , Receptors, Chemokine/metabolism , Severity of Illness Index
14.
Curr Drug Targets Inflamm Allergy ; 2(4): 313-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14561150

ABSTRACT

We review evidence that Stem Cell Factor (SCF) plays an important role in the pathophysiology of asthma. SCF is produced by a wide variety of cells present in asthmatic lung, including mast cells and eosinophils. Its receptor, c-kit, is broadly expressed on mature mast cells and eosinophils. SCF promotes recruitment of mast cell progenitors into tissues, as well as their local maturation and activation. It also promotes eosinophil survival, maturation and functional activation. SCF enhances IgE-dependent release of mediators from mast cells, including histamine, leukotrienes, cytokines (TNF-alpha, IL-5, GM-CSF) and chemokines (RANTES/CCL5, MCP-1/CCL2, TARC/CCL17 e MDC/CCL22); it is required for IL-4 production in mast cells. SCF, acting in concert with IgE, also upregulates the expression and function of CC chemokine receptors in mast cells. Structural and resident airway cells express increased levels of SCF in the bronchus of asthmatic patients. In a murine model of asthma, allergen exposure increased production of SCF by epithelial cells and alveolar macrophages, which was transient and paralleled by histamine release. SCF induced long-lived airway hyperreactivity, which was prevented by local neutralization of SCF, as well as by inhibitors of the production or activity of cysteinyl-leukotrienes. Together, these observations suggest that SCF has an important role in asthma.


Subject(s)
Asthma/physiopathology , Cytokines/physiology , Hematopoiesis/physiology , Stem Cell Factor/physiology , Drug Delivery Systems , Humans , Mast Cells/physiology
15.
J Neurosci Res ; 66(4): 705-14, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11746391

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a CD4(+) T cell disease of the central nervous system (CNS) characterized by mononuclear cell infiltration, demyelination, and paralysis. Recent studies describing the relationship of chemokine expression with development of clinical disease have led to the hypothesis that distinct chemokine receptors corresponding to specific ligands are expressed by CNS-infiltrating antigen-specific encephalitogenic T cells as well as host-derived bystander T cells and monocytes. In an effort to study encephalitogenic T cell chemokine receptor expression, we examined CC chemokine receptor expression from resting, activated, and CNS-isolated CD4(+) T cells. CCR1, CCR2, CCR3, CCR5, CCR6, CCR7, and CCR8 mRNA is expressed by normal CD4(+) T cells. In vitro activated T cells expressed CCR1, CCR2, CCR3, CCR5, CCR6, CCR7, and CCR8 mRNA as well as CCR4. After EAE induction, CCR1 mRNA was expressed by donor-derived encephalitogenic and host-derived CD4(+) T cells isolated only from CNS and not from spleen. In vivo neutralization of the CCR1 ligand, macrophage inflammatory protein-1alpha (CCL3), resulted in less encephalitogenic CD4(+) T cell CNS infiltration. These results demonstrate the importance of CC chemokine receptor expression by CD4(+) encephalitogenic T cells for CNS infiltration and subsequent disease development.


Subject(s)
Central Nervous System/immunology , Chemotaxis, Leukocyte/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gene Expression Regulation/immunology , Receptors, Chemokine/genetics , T-Lymphocytes/immunology , Animals , Antibodies/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Central Nervous System/metabolism , Central Nervous System/physiopathology , Chemokine CCL3 , Chemokine CCL4 , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Interferon-gamma/genetics , Interleukin-4/genetics , Macrophage Inflammatory Proteins/antagonists & inhibitors , Macrophage Inflammatory Proteins/immunology , Mice , Mice, Congenic , RNA, Messenger/immunology , RNA, Messenger/metabolism , Receptors, CCR1 , Receptors, CCR2 , Receptors, CCR3 , Receptors, CCR4 , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Recurrence , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Thy-1 Antigens/genetics , Thy-1 Antigens/immunology
16.
J Immunol ; 167(8): 4635-43, 2001 Oct 15.
Article in English | MEDLINE | ID: mdl-11591793

ABSTRACT

Macrophage-inflammatory protein 2 (MIP-2) is a major CXC chemokine involved in the migration of polymorphonuclear neutrophils (PMNs) to sites of inflammation. Although cell culture experiments have identified different cell types that can produce MIP-2, the cellular sources in vivo are not clearly defined. By using immunohistochemical staining and analysis of chemokine mRNA expression, the present study aimed to localize cells producing MIP-2 in tissues of normal mice and mice challenged with Yersinia enterocolitica. The results showed a constitutive expression of MIP-2 mRNA in bone marrow (BM) of normal mice, but not in other organs such as spleen, lung, or liver. MIP-2 protein was found in all organs tested but it was exclusively associated with PMNs that stained positive with the cell surface marker Gr-1. Bacterial infection caused a 5-fold increase in the number of MIP-2-positive PMNs recruited to spleens concomitant with a strong increase of splenic MIP-2 mRNA. This correlated well with a 3-fold loss of MIP-2-producing cells in BM. Because MIP-2 mRNA expression in PMNs was increased after stimulation with TNF, the results indicate that newly recruited PMNs can supplement their MIP-2 content through TNF-stimulated transcription. Together, the data imply a constitutive production of MIP-2 by a subset of PMNs in BM and argue for the possibility of a rapid mobilization of MIP-2 through its storage in circulating PMNs.


Subject(s)
Bone Marrow Cells/chemistry , Chemokines/isolation & purification , Chemotaxis, Leukocyte , Leukopoiesis , Neutrophils/chemistry , Animals , Bone Marrow Cells/immunology , Chemokine CXCL2 , Chemokines/biosynthesis , Chemokines/genetics , Female , Granulocytes/chemistry , Interleukin-1/pharmacology , Mice , Mice, Inbred BALB C , Neutrophils/immunology , RNA, Messenger/isolation & purification , Spleen/cytology , Spleen/immunology , Tissue Distribution , Tumor Necrosis Factor-alpha/pharmacology , Yersinia Infections/immunology , Yersinia enterocolitica
17.
Am J Pathol ; 159(3): 1171-80, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11549610

ABSTRACT

Thrombin, an important clotting factor, extravasates at sites of blood-retina barrier breakdown that is often associated with many retinal diseases. Here we investigated the effects of thrombin on human retinal pigment epithelial (HRPE) cells, monocytes, and HRPE cell/monocyte co-cultures. Thrombin induced secretion and mRNA expression of HRPE interleukin (IL)-8 and monocyte chemoattractant protein-1 (MCP-1). Thrombin also enhanced IL-8 and MCP-1 by HRPE cell/monocyte co-cultures, by apparently enhancing cell-cell contact mechanisms. The thrombin effects on IL-6 secretion were similar to those on chemokine secretion. Thrombin-induced chemokines by co-cultures were inhibited by anti-tumor necrosis factor-alpha (TNF-alpha) antibody, but not by anti-IL-1beta antibody. TNF-alpha was detected in cell lysates of monocytes detached from HRPE cells after co-culture stimulation with thrombin. HRPE cells mainly produced these chemokines. However, thrombin generally potentiated exogenous IL-1beta- and TNF-alpha-induced chemokine production by HRPE cells, monocytes, and co-cultures. Interferon-gamma potentiated chemokine secretion by co-cultures with or without thrombin. Our results indicate that thrombin may cause leukocyte recruitment by inducing HRPE cell and monocyte chemokine and by enhancing HRPE cell/monocyte interactions, in part because of monocyte TNF-alpha induction, suggesting important mechanisms for ocular inflammation during blood-retina barrier breakdown and intra-ocular hemorrhage.


Subject(s)
Cell Communication/physiology , Chemokines/metabolism , Monocytes/physiology , Pigment Epithelium of Eye/physiology , Thrombin/physiology , Antibodies/pharmacology , Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/metabolism , Coculture Techniques , Humans , Interferon-gamma/pharmacology , Interleukin-1/pharmacology , Interleukin-6/metabolism , Interleukin-8/antagonists & inhibitors , Interleukin-8/metabolism , Pigment Epithelium of Eye/cytology , Recombinant Proteins , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology
18.
J Exp Med ; 194(4): 551-5, 2001 Aug 20.
Article in English | MEDLINE | ID: mdl-11514610

ABSTRACT

Allergic asthmatic responses in the airway are associated with airway hyperreactivity, eosinophil accumulation in the lung, and cytokine production by allergen-specific, T helper cell type 2 (Th2) lymphocytes. Here, we show that in a cockroach antigen (CA) model of allergic pulmonary inflammation, the chemokine macrophage inflammatory protein (MIP)-3alpha is expressed in the lung within hours of allergen challenge. To determine the biologic relevance of this expression, mice lacking CCR6, the only known receptor for MIP-3alpha, were studied for their response to CA. CCR6-deficient mice were immunized to the same extent as their wild-type counterparts, as judged by cytokine production in antigen-challenged lymphocytes. However, compared with CA-challenged wild-type mice, challenged CCR6-deficient mice had reduced airway resistance, fewer eosinophils around the airway, lower levels of interleukin 5 in the lung, and reduced serum levels of immunoglobulin E. Together, these data demonstrate that MIP-3alpha and CCR6 function in allergic pulmonary responses and suggest that these molecules might represent novel therapeutic targets for treatment of asthma.


Subject(s)
Asthma/physiopathology , Hypersensitivity/physiopathology , Pneumonia/physiopathology , Receptors, Chemokine/physiology , Animals , Asthma/immunology , Asthma/metabolism , Cytokines/metabolism , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunoglobulin E/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/immunology , Pneumonia/metabolism , Receptors, CCR6 , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
19.
Am J Pathol ; 159(2): 631-8, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11485921

ABSTRACT

Respiratory syncytial virus (RSV) is a respiratory pathogen that can cause significant morbidity in infants and young children. Interestingly, the majority of children who acquire a RSV infection do not exhibit severe symptoms. Development of a Th1 response has been associated with resolution of symptoms in viral infections and may explain mild RSV illness. The current study investigated the cytokine response observed in mild disease in C57BL/6 mice that had low airway resistance and mucus production with little pulmonary inflammation. RSV infection in these mice was accompanied by a fourfold increase in interleukin-12(IL-12). Treatment of RSV-infected mice with anti-IL-12 resulted in an increase in airway hyperreactivity, mucus production, and airway inflammation (eosinophilia). Since IL-12 activation is dependent on Stat-4-mediated intracellular signal transduction, similar experiments were performed in Stat-4 deficient mice and demonstrated similar results to those obtained from anti-IL-12 treated mice. Again, there was an increase in airway hyperreactivity and mucus production, and goblet cell hypertrophy. These studies support the importance of IL-12 in the immune response to RSV infection resulting in resolution of disease and protection from inappropriate inflammatory responses.


Subject(s)
Bronchial Hyperreactivity/physiopathology , DNA-Binding Proteins/physiology , Interleukin-12/physiology , Lung/pathology , Respiratory Syncytial Virus Infections/physiopathology , Trans-Activators/physiology , Animals , Antibodies/pharmacology , Bronchoalveolar Lavage Fluid/cytology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Eosinophils/physiology , Inflammation/physiopathology , Interleukin-12/immunology , Interleukin-13/biosynthesis , Lung/physiopathology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , STAT4 Transcription Factor , Signal Transduction , Time Factors , Trans-Activators/deficiency , Trans-Activators/genetics
20.
J Immunol ; 167(2): 1060-5, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11441116

ABSTRACT

The development of severe childhood asthma may be influenced by several factors including environmental and infectious stimuli. The causal relationship between infectious viral responses, such as respiratory syncytial virus (RSV), and severe asthma during early childhood is unclear. In these studies, the ability for an initial RSV infection to exacerbate and promote a more severe asthmatic-type response was investigated by combining established murine models of disease. We examined the ability of RSV to induce exacerbation of allergic disease over a relatively long period, leading to development of severe airway responses including airway inflammation and hyperreactivity. The preferential production of IL-13 during a primary RSV infection appears to play a critical role for the exacerbation of cockroach allergen-induced disease. The depletion of IL-13 during RSV infections inhibited the exacerbation and acceleration of severe allergen-induced airway hyperreactivity. This was indicated by decreases in airway hyperreactivity and changes in lung chemokine production. These data suggest that the airway responses during asthma can be greatly affected by a previous RSV infection, even when infection occurs before allergen sensitization. Overall, infection of the airways with RSV can induce an IL-13-dependent change in airway function and promotes an environment that contributes to the development of severe allergic asthmatic responses.


Subject(s)
Interleukin-13/physiology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/virology , Respiratory Syncytial Virus, Human/immunology , Allergens/immunology , Animals , Antigens, Plant , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/prevention & control , Bronchial Hyperreactivity/virology , Chemokines/biosynthesis , Cockroaches/immunology , Cytokines/biosynthesis , Immune Sera/administration & dosage , Injections, Intraperitoneal , Interleukin-13/antagonists & inhibitors , Interleukin-13/immunology , Lung/immunology , Lung/metabolism , Mice , Mice, Inbred BALB C , Respiratory Hypersensitivity/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...