Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Immunology ; 152(1): 138-149, 2017 09.
Article in English | MEDLINE | ID: mdl-28502093

ABSTRACT

The Toll-like receptor (TLR) adaptor proteins myeloid differentiating factor 88 (MyD88) and Toll, interleukin-1 receptor and resistance protein (TIR) domain-containing adaptor inducing interferon-ß (TRIF) comprise the two principal limbs of the TLR signalling network. We studied the role of these adaptors in the TLR4-dependent inhibition of allergic airway disease and induction of CD4+ ICOS+ T cells by nasal application of Protollin™, a mucosal adjuvant composed of TLR2 and TLR4 agonists. Wild-type (WT), Trif-/- or Myd88-/- mice were sensitized to birch pollen extract (BPEx), then received intranasal Protollin followed by consecutive BPEx challenges. Protollin's protection against allergic airway disease was TRIF-dependent and MyD88-independent. TRIF deficiency diminished the CD4+ ICOS+ T-cell subsets in the lymph nodes draining the nasal mucosa, as well as their recruitment to the lungs. Overall, TRIF deficiency reduced the proportion of cervical lymph node and lung CD4+ ICOS+ Foxp3- cells, in particular. Adoptive transfer of cervical lymph node cells supported a role for Protollin-induced CD4+ ICOS+ cells in the TRIF-dependent inhibition of airway hyper-responsiveness. Hence, our data demonstrate that stimulation of the TLR4-TRIF pathway can protect against the development of allergic airway disease and that a TRIF-dependent adjuvant effect on CD4+ ICOS+ T-cell responses may be a contributing mechanism.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Asthma/prevention & control , CD4-Positive T-Lymphocytes/metabolism , Lung/metabolism , Rhinitis, Allergic, Seasonal/prevention & control , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/immunology , Adoptive Transfer , Animals , Antigens, Plant/immunology , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , Betula/immunology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/metabolism , Bronchial Hyperreactivity/physiopathology , Bronchial Hyperreactivity/prevention & control , Bronchoconstriction , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/transplantation , Cell Proliferation , Chemotaxis, Leukocyte , Cysteine Endopeptidases/immunology , Disease Models, Animal , Drug Combinations , Female , Genetic Predisposition to Disease , Inducible T-Cell Co-Stimulator Protein/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Lipopolysaccharides/immunology , Lung/immunology , Lung/physiopathology , Lymphocyte Activation , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Phenotype , Pollen/immunology , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/metabolism , Rhinitis, Allergic, Seasonal/physiopathology , Signal Transduction , Time Factors , Toll-Like Receptor 4/immunology
2.
Physiol Plant ; 159(4): 468-482, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27859326

ABSTRACT

The main factors regulating grapevine response to decreasing water availability were assessed under statistical support using published data related to leaf water relations in an extensive range of scion and rootstock genotypes. Matching leaf water potential (Ψleaf ) and stomatal conductance (gs ) data were collected from peer-reviewed literature with associated information. The resulting database contained 718 data points from 26 different Vitis vinifera varieties investigated as scions, 15 non-V. vinifera rootstock genotypes and 11 own-rooted V. vinifera varieties. Linearised data were analysed using the univariate general linear model (GLM) with factorial design including biological (scion and rootstock genotypes), methodological and environmental (soil) fixed factors. The first GLM performed on the whole database explained 82.4% of the variability in data distribution having the rootstock genotype the greatest contribution to variability (19.1%) followed by the scion genotype (16.2%). A classification of scions and rootstocks according to their mean predicted gs in response to moderate water stress was generated. This model also revealed that gs data obtained using a porometer were in average 2.1 times higher than using an infra-red gas analyser. The effect of soil water-holding properties was evaluated in a second analysis on a restricted database and showed a scion-dependant effect, which was dominant over rootstock effect, in predicting gs values. Overall the results suggest that a continuum exists in the range of stomatal sensitivities to water stress in V. vinifera, rather than an isohydric-anisohydric dichotomy, that is further enriched by the diversity of scion-rootstock combinations and their interaction with different soils.


Subject(s)
Plant Stomata/physiology , Vitis/physiology , Water/physiology , Databases as Topic , Dehydration , Linear Models , Models, Biological , Soil
3.
J Immunol ; 194(12): 5626-34, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25934863

ABSTRACT

Contact between airway smooth muscle (ASM) cells and activated CD4(+) T cells, a key interaction in diseases such as asthma, triggers ASM cell proliferation and enhances T cell survival. We hypothesized that direct contact between ASM and CD4(+) T cells facilitated the transfer of anti-apoptotic proteins via nanotubes, resulting in increased survival of activated CD4(+) T cells. CD4(+) T cells, isolated from PBMCs of healthy subjects, when activated and cocultured with ASM cells for 24 h, formed nanotubes that were visualized by immunofluorescence and atomic force microscopy. Cell-to-cell transfer of the fluorescent dye calcein-AM confirmed cytoplasmic communication via nanotubes. Immunoreactive B cell lymphoma 2 (Bcl-2) and induced myeloid leukemia cell differentiation protein (Mcl-1), two major anti-apoptotic proteins, were present within the nanotubes. Downregulation of Mcl-1 by small interfering RNA in ASM cells significantly increased T cell apoptosis, whereas downregulation of Bcl-2 had no effect. Transfer of GFP-tagged Mcl-1 from ASM cells to CD4(+) T cells via the nanotubes confirmed directionality of transfer. In conclusion, activated T cells communicate with ASM cells via nanotube formation. Direct transfer of Mcl-1 from ASM to CD(+) T cells via nanotubes is involved in T cell survival. This study provides a novel mechanism of survival of CD4(+) T cells that is dependent on interaction with a structural cell.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Myocytes, Smooth Muscle/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Apoptosis/immunology , Biological Transport , CD4-Positive T-Lymphocytes/drug effects , Calcium/metabolism , Calcium Signaling , Cell Adhesion/immunology , Cell Communication/immunology , Cell Survival/immunology , Cells, Cultured , Coculture Techniques , Humans , Hyaluronan Receptors/immunology , Lymphocyte Activation/immunology
4.
Hum Mol Genet ; 21(10): 2277-87, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22343142

ABSTRACT

Nutrient-deprivation autophagy factor-1 (NAF-1) was identified as an endoplasmic reticulum (ER) BCL-2-interacting protein, which functions to mediate the ability of ER BCL-2 to antagonize Beclin 1-dependent autophagy and depress ER calcium stores. In humans, a point mutation in Naf-1 (synonyms: Cisd2, Eris, Miner1 and Noxp70) is responsible for the neurodegenerative disorder Wolfram Syndrome 2. Here, we describe the generation and characterization of the Naf-1 gene deletion in mice. Naf-1 null mice display discernable clinical signs of degeneration at 2-3 months of age, with early evidence of significant defects in the structure and performance of skeletal muscle. Skeletal muscles from Naf-1 knockout mice demonstrate a significant shift towards slow-twitch (type I) fibers and greater resistance to muscle fatigue. Force-generating capacity is dramatically reduced in Naf-1(-/-) muscle. Consistent with its role in ER BCL-2-mediated regulation of autophagy and calcium flux, these physiological deficiencies were accompanied by augmented autophagy and dysregulated calcium homeostasis. In contrast, this also included adaptive enlargement of mitochondria with extensive cristae structures. Thus, NAF-1, a BCL-2-associated autophagy regulator, is required for homeostatic maintenance of skeletal muscle. Our findings uncover a novel pathway that is required for normal muscle maintenance, which may ultimately provide a novel therapeutic target for treating certain muscle pathologies.


Subject(s)
Autophagy , Carrier Proteins/genetics , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/genetics , Proto-Oncogene Proteins/metabolism , Ribonucleoproteins/genetics , Animals , Autophagy-Related Proteins , Carrier Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2 , Ribonucleoproteins/metabolism
5.
Am J Respir Cell Mol Biol ; 49(4): 563-70, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23656597

ABSTRACT

Allergic asthma is a heterogeneous disease with no curative therapies. T cells infiltrate the airway smooth muscle (ASM) layer and may be implicated in airway remodeling and the increase of ASM mass, a cardinal feature of asthma. The mechanism by which CD4(+) T cells drive airway remodeling remains unknown. This study sought to determine the T cell-mediated mechanism of ASM cell proliferation. We hypothesized that CD4(+) T cells adhere to ASM cells via CD44, and induce ASM cell proliferation through the activation of the epidermal growth factor receptor (EGFR). A coculture model showed that the contact of antigen-stimulated CD4(+) T cells with ASM cells induced high levels of EGFR ligand expression in CD4(+) T cells and the activation of matrix metalloproteinase (MMP)-9, required for the shedding of EGFR ligands. The inhibition of EGFR and MMP-9 prevented the increase of ASM cell proliferation after coculture. The hyaluronan receptor CD44 is the dominant mediator of the tight adherence of T cells to ASM and is colocalized with MMP-9 on the cell surface. Moreover, the neutralization of CD44 prevents ASM cell hyperplasia. These data provide a novel mechanism by which antigen-stimulated CD4(+) T cells induce the remodeling indicative of a direct trophic role for CD4(+) T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , ErbB Receptors/metabolism , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/metabolism , Respiratory System/immunology , Respiratory System/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Adhesion/immunology , Cells, Cultured , Coculture Techniques , ErbB Receptors/immunology , Hyaluronan Receptors/immunology , Lymphocyte Activation , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Muscle, Smooth/immunology , Myocytes, Smooth Muscle/immunology , Rats
6.
Respir Res ; 14: 118, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24283210

ABSTRACT

BACKGROUND: Allergic asthma is characterized by airway inflammation in response to antigen exposure, leading to airway remodeling and lung dysfunction. Epithelial-mesenchymal transition (EMT) may play a role in airway remodeling through the acquisition of a mesenchymal phenotype in airway epithelial cells. TGF-ß1 is known to promote EMT; however, other cytokines expressed in severe asthma with extensive remodeling, such as IL-22, may also contribute to this process. In this study, we evaluated the contribution of IL-22 to EMT in primary bronchial epithelial cells from healthy and asthmatic subjects. METHODS: Primary bronchial epithelial cells were isolated from healthy subjects, mild asthmatics and severe asthmatics (n=5 patients per group). The mRNA and protein expression of epithelial and mesenchymal cell markers and EMT-associated transcription factors was evaluated following stimulation with TGF-ß1, IL-22 and TGF-ß1+IL-22. RESULTS: Primary bronchial epithelial cells stimulated with TGF-ß1 underwent EMT, demonstrated by decreased expression of epithelial markers (E-cadherin and MUC5AC) and increased expression of mesenchymal markers (N-cadherin and vimentin) and EMT-associated transcription factors. IL-22 alone had no effect on epithelial or mesenchymal gene expression. However, IL-22+TGF-ß1 promoted the expression of some EMT transcription factors (Snail1 and Zeb1) and led to a more profound cadherin shift, but only in cells obtained from severe asthmatics. CONCLUSION: The impact of IL-22 on airway epithelial cells depends on the cytokine milieu and the clinical phenotype of the patient. Further studies are required to determine the molecular mechanism of IL-22 and TGF-ß1 cooperativity in driving EMT in primary human bronchial epithelial cells.


Subject(s)
Asthma/physiopathology , Bronchi/physiopathology , Epithelial Cells/physiology , Epithelial-Mesenchymal Transition/physiology , Interleukins/physiology , Transforming Growth Factor beta1/physiology , Adolescent , Adult , Aged , Asthma/metabolism , Asthma/pathology , Biopsy , Bronchi/drug effects , Bronchi/pathology , Cadherins/metabolism , Case-Control Studies , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , In Vitro Techniques , Interleukins/pharmacology , Male , Middle Aged , Mucin 5AC/metabolism , Phenotype , RNA, Messenger/metabolism , Severity of Illness Index , Transforming Growth Factor beta1/pharmacology , Young Adult , Interleukin-22
7.
FASEB J ; 26(4): 1704-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22247333

ABSTRACT

Asthma is a chronic inflammatory disease that is associated with airway remodeling, including hyperplasia of airway epithelial cells and airway smooth muscle cells, and goblet cell differentiation. We wished to address the potential role of histamine, a key biogenic amine involved in allergic reactions, in airway remodeling through the epidermal growth factor receptor (EGFR) pathway. Here, we demonstrate that histamine releases 2 EGFR ligands, amphiregulin and heparin-binding epidermal growth factor-like growth factor (HB-EGF), from airway epithelial cells. Amphiregulin and HB-EGF were expressed in airway epithelium of patients with asthma. Histamine up-regulated their mRNA expression (amphiregulin 3.2-fold, P<0.001; HB-EGF 2.3-fold, P<0.05) and triggered their release (amphiregulin EC(50) 0.50 µM, 31.2 ± 2.7 pg/ml with 10 µM histamine, P<0.01; HB-EGF EC(50) 0.54 µM, 78.5 ± 1.8 pg/ml with 10 µM histamine, P<0.001) compared to vehicle control (amphiregulin 19.3 ± 0.9 pg/ml; HB-EGF 60.2 ± 1.0 pg/ml), in airway epithelial cells. Histamine increased EGFR phosphorylation (2.1-fold by Western blot analysis) and induced goblet cell differentiation (CLCA1 up-regulation by real-time qPCR) in normal human bronchial epithelial (NHBE) cells. Moreover, amphiregulin and HB-EGF caused proliferation and migration of both NHBE cells and human airway smooth muscle cells. These results suggest that histamine may induce airway remodeling via the epithelial-derived EGFR ligands amphiregulin and HB-EGF.


Subject(s)
Airway Remodeling/drug effects , Bronchi/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Histamine/pharmacology , Adult , Amphiregulin , Asthma/metabolism , Asthma/pathology , Cell Differentiation/drug effects , Cell Line , Cell Movement , Cell Proliferation , EGF Family of Proteins , Epithelial Cells/cytology , Glycoproteins/genetics , Glycoproteins/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Middle Aged , Receptors, Histamine H1/metabolism , Young Adult
8.
FASEB J ; 26(12): 5152-60, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22898922

ABSTRACT

Increased airway smooth muscle (ASM) mass is a hallmark of airway remodeling in severe asthma. Th17-associated cytokines, particularly IL-17A, IL-17F, and IL-22, have been postulated to play a role in the pathogenesis of asthma. To investigate the in vitro effect of Th17 cytokines on the proliferation and survival of airway smooth muscle cells (ASMCs), human ASMCs from asthmatic and nonasthmatic subjects were incubated with IL-17A, IL-17F, or IL-22. The aforementioned cytokines demonstrated an ability to promote proliferation and survival of ASMCs from asthmatic and nonasthmatic subjects, which were mediated by selective activation of their corresponding receptors on ASMCs, including IL-17RA, IL-17RC, or IL-22R1, respectively. IL-17A and IL-17F-induced proliferation of ASMCs was dependent on ERK1/2 MAPK pathway, while IL-22-induced proliferation involved both ERK1/2 MAPK and NF-κB pathways. The involvement of signaling pathways was further confirmed by the inhibition of proliferation by knockdown of ERK1/2 MAPK or NF-κB p65 expression with pathway-specific siRNA. Together, our results show that Th17-associated cytokines promote proliferation and reduce the apoptotic rate of human ASMCs, raising the possibility that Th17 cytokines may contribute to increasing airway smooth muscle mass and airway remodeling in asthma.


Subject(s)
Cell Proliferation/drug effects , Cytokines/pharmacology , Myocytes, Smooth Muscle/drug effects , Th17 Cells/metabolism , Apoptosis/drug effects , Asthma/metabolism , Asthma/pathology , Benzamides/pharmacology , Bronchi/metabolism , Bronchi/pathology , Cell Survival/drug effects , Cells, Cultured , Humans , Immunohistochemistry , Interleukin-17/pharmacology , Interleukins/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , RNA Interference , Receptors, Interleukin/metabolism , Receptors, Interleukin-17/metabolism , Sesquiterpenes/pharmacology , Sesquiterpenes, Guaiane , Signal Transduction/drug effects , Interleukin-22
9.
Pulm Pharmacol Ther ; 26(1): 13-23, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22561160

ABSTRACT

Smooth muscle comprises a key functional component of both the airways and their supporting vasculature. Dysfunction of smooth muscle contributes to and exacerbates a host of breathing-associated pathologies such as asthma, chronic obstructive pulmonary disease and pulmonary hypertension. These diseases may be marked by airway and/or vascular smooth muscle hypertrophy, proliferation and hyper-reactivity, and related conditions such as fibrosis and extracellular matrix remodeling. This review will focus on the contribution of airway or vascular smooth dysfunction to common airway diseases.


Subject(s)
Asthma/physiopathology , Hypertension, Pulmonary/physiopathology , Muscle, Smooth/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Airway Remodeling , Animals , Bronchial Hyperreactivity/physiopathology , Cell Proliferation , Humans , Hypertrophy , Muscle, Smooth/cytology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology
10.
Pulm Pharmacol Ther ; 26(1): 24-36, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22967819

ABSTRACT

Asthma is a chronic obstructive airway disease characterised by airway hyperresponsiveness (AHR) and airway wall remodelling. The effector of airway narrowing is the contraction of airway smooth muscle (ASM), yet the question of whether an inherent or acquired dysfunction in ASM contractile function plays a significant role in the disease pathophysiology remains contentious. The difficulty in determining the role of ASM lies in limitations with the models used to assess contraction. In vivo models provide a fully integrated physiological response but ASM contraction cannot be directly measured. Ex vivo and in vitro models can provide more direct assessment of ASM contraction but the loss of factors that may modulate ASM responsiveness and AHR, including interaction between multiple cell types and disruption of the mechanical environment, precludes a complete understanding of the disease process. In this review we detail key advantages of common in vivo, ex vivo and in vitro models of ASM contraction, as well as emerging tissue engineered models of ASM and whole airways. We also highlight important findings from each model with respect to the pathophysiology of asthma.


Subject(s)
Asthma/physiopathology , Models, Biological , Muscle, Smooth/metabolism , Animals , Bronchial Hyperreactivity/physiopathology , Disease Models, Animal , Humans , Muscle Contraction/physiology , Tissue Engineering
11.
J Allergy Clin Immunol ; 130(4): 977-85.e6, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22698519

ABSTRACT

BACKGROUND: Airway smooth muscle cell (ASMC) migration is one of the proposed mechanisms underlying the increased airway smooth muscle mass seen in airway remodeling of patients with severe asthma. IL-17-related cytokines are a new subgroup of inflammatory mediators that have been suggested to play a role in regulating smooth muscle function. We hypothesized that IL-17-induced chemokine production from smooth muscle cells can contribute to migration of additional smooth muscle cells in the airways of asthmatic patients. OBJECTIVE: We sought to investigate the effect of IL-17 on smooth muscle-derived chemokines and to examine the mechanisms involved in their production and contribution to the increase in airway smooth muscle migration. METHODS: The effect of IL-17-induced supernatants on human ASMC migration was investigated. IL-17-induced growth-related oncogene (GRO) production and mRNA expression was assessed by using ELISA and RT-PCR, respectively. The direct effect of GROs on ASMC migration and the involvement of the CXCR2 receptor were also examined. RESULTS: IL-17-induced supernatants promoted ASMC migration. After IL-17 stimulation, GROs were the most abundant chemokines produced from ASMCs, and blocking their effect by using neutralizing antibodies significantly inhibited ASMC migration. In addition, a combination of recombinant human GRO-α, GRO-ß, and GRO-γ was able to promote significant migration of ASMCs that was mediated through the CXCR2 receptor. CONCLUSION: These findings suggest that IL-17-induced GROs can be an important mediator of ASMC migration and therefore might contribute to the pathogenesis of airway remodeling in asthmatic patients.


Subject(s)
Autocrine Communication , Bronchi/cytology , Cell Movement , Chemokine CXCL1/physiology , Interleukin-17/pharmacology , Myocytes, Smooth Muscle/physiology , Cell Movement/drug effects , Chemokine CXCL1/genetics , Extracellular Signal-Regulated MAP Kinases/physiology , Humans , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Receptors, Interleukin-8B/physiology , p38 Mitogen-Activated Protein Kinases/physiology
12.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L54-63, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22523281

ABSTRACT

Ovalbumin (OVA) is the most frequently used allergen in animal models of asthma. Lipopolysaccharide (LPS) contaminating commercial OVA may modulate the evoked airway inflammatory response to OVA. However, the effect of LPS in OVA on airway remodeling, especially airway smooth muscle (ASM) has not been evaluated. We hypothesized that LPS in commercial OVA may enhance allergen-induced airway inflammation and remodeling. Brown Norway rats were sensitized with OVA on day 0. PBS, OVA, or endotoxin-free OVA (Ef-OVA) was instilled intratracheally on days 14, 19, 24. Bronchoalveolar lavage (BAL) fluid, lung, and intrathoracic lymph node tissues were collected 48 h after the last challenge. Immunohistochemistry for α-smooth muscle actin, Periodic-Acid-Schiff staining, and real-time qPCR were performed. Airway hyperresponsiveness (AHR) was also measured. BAL fluid macrophages, eosinophils, neutrophils, and lymphocytes were increased in OVA-challenged animals, and macrophages and neutrophils were significantly lower in Ef-OVA-challenged animals. The ASM area in larger airways was significantly increased in both OVA and Ef-OVA compared with PBS-challenged animals. The mRNA expression of IFN-γ and IL-13 in lung tissues and IL-4 in lymph nodes was significantly increased by both OVA and Ef-OVA compared with PBS and were not significantly different between OVA and Ef-OVA. Monocyte chemoattractant protein (MCP)-1 in BAL fluid and AHR were significantly increased in OVA but not in Ef-OVA. LPS contamination in OVA contributes to the influx of macrophages and MCP-1 increase in the airways and to AHR after OVA challenges but does not affect OVA-induced Th1 and Th2 cytokine expression, goblet cell hyperplasia, and ASM remodeling.


Subject(s)
Airway Remodeling/drug effects , Airway Remodeling/immunology , Asthma/immunology , Inflammation/immunology , Lipopolysaccharides/immunology , Ovalbumin/immunology , Animals , Asthma/chemically induced , Asthma/metabolism , Asthma/pathology , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Bronchoalveolar Lavage Fluid/immunology , Chemokine CCL2/immunology , Chemokine CXCL1/immunology , Disease Models, Animal , Drug Contamination , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/metabolism , Eosinophils/pathology , ErbB Receptors/immunology , ErbB Receptors/metabolism , Hyperplasia/immunology , Hyperplasia/pathology , Inflammation/chemically induced , Inflammation/pathology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , Lipopolysaccharides/pharmacology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Muscle, Smooth/drug effects , Muscle, Smooth/immunology , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , NF-kappa B/immunology , NF-kappa B/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Ovalbumin/pharmacology , Rats , Rats, Inbred BN
13.
Am J Physiol Lung Cell Mol Physiol ; 302(8): L736-45, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22287614

ABSTRACT

Sphingosine-1-phosphate (S1P) is an immunomodulatory lipid mediator that plays an important role in lymphocyte trafficking. Elevated levels of S1P are found in bronchoalveolar lavage (BAL) fluid of patients with asthma; however, its role in disease is not known. FTY720, a synthetic analog of S1P, has been shown to abrogate allergic inflammation and airway hyperresponsiveness following acute allergen challenge. However, its effects on asthmatic airway remodeling induced by repeated allergen exposure are unknown. Ovalbumin (OVA)-sensitized rats were challenged on days 14, 19, and 24 after sensitization. FTY720 or vehicle (PBS) therapy was administered 1 h prior to each challenge. BAL fluid and quantitative histological analysis were performed 48 h after the last challenge. FTY720 inhibited OVA-induced features of airway remodeling including increased airway smooth muscle mass and bronchial neovascularization, without affecting lymphocyte numbers in secondary lymphoid organs. Furthermore, CD3+ cells adjacent to airway smooth muscle bundles were increased in OVA-challenged rats but the increase was inhibited by FTY720. There was an expansion of bronchus-associated lymphoid tissue following FTY720 treatment of OVA-challenged animals. Real-time quantitative PCR revealed that Th2-associated transcription factors were inhibited following FTY720 therapy. Airway remodeling is a cardinal feature of severe asthma. These results demonstrate that allergen-driven airway remodeling can be inhibited by FTY720, offering potential new therapies for the treatment of severe asthma.


Subject(s)
Airway Remodeling/drug effects , Allergens/adverse effects , Immunosuppressive Agents/therapeutic use , Lysophospholipids/agonists , Lysophospholipids/therapeutic use , Propylene Glycols/therapeutic use , Sphingosine/analogs & derivatives , Animals , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Bronchi/blood supply , Bronchi/drug effects , Bronchi/pathology , Bronchoalveolar Lavage Fluid , CD3 Complex/analysis , Fingolimod Hydrochloride , Male , Muscle, Smooth/drug effects , Rats , Severity of Illness Index , Sphingosine/agonists , Sphingosine/therapeutic use , Treatment Outcome
14.
J Allergy Clin Immunol ; 127(4): 1046-53.e1-2, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21345484

ABSTRACT

BACKGROUND: Migration of airway smooth muscle cells (ASMCs) might contribute to increased airway smooth muscle mass in asthma. T(H)17 cells and T(H)17-associated cytokines are involved in the pathogenesis of asthma and might also contribute to airway remodeling. OBJECTIVE: We sought to explore the possibility that migration of ASMCs might contribute to airway remodeling through the action of T(H)17-related cytokines. METHODS: The effect of exogenous T(H)17 cytokines on ex vivo human ASMC migration was investigated by using a chemotaxis assay. The involvement of signaling pathways, including p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 1/2 MAPK, nuclear factor κB, and phosphoinositide 3-kinase, was also examined. RESULTS: We demonstrated that IL-17A, IL-17F, and IL-22 promote migration in a dose-dependent manner. We further demonstrated that ASMCs express receptors for IL-17RA, IL-17RC, and IL-22R1. Using mAbs directed against these receptors, we confirmed that T(H)17-associated cytokine-induced migration was dependent on selective receptor activation. Moreover, IL-17A and IL-17F exert their effects through signaling pathways that are distinct from those used by IL-22. The p38 MAPK inhibitor BIRB0796 inhibited the migration induced by IL-17A and IL-17F. PS1145, an inhibitor of nuclear factor κB, abolished the IL-22-induced migration. CONCLUSION: These data raise the possibility that T(H)17-associated cytokines promote human ASMC migration in vivo and suggest an important new mechanism for the promotion of airway remodeling in asthma.


Subject(s)
Airway Remodeling/physiology , Cell Movement/physiology , Cytokines/metabolism , Myocytes, Smooth Muscle/cytology , Th17 Cells/metabolism , Asthma/immunology , Asthma/metabolism , Asthma/physiopathology , Cell Separation , Cytokines/immunology , Flow Cytometry , Humans , Myocytes, Smooth Muscle/metabolism , Signal Transduction/physiology , Th17 Cells/immunology
15.
Am J Respir Cell Mol Biol ; 45(5): 984-90, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21493783

ABSTRACT

Severe or refractory asthma affects 5 to 15% of all patients with asthma, but is responsible for more than half of the health burden associated with the disease. Severe asthma is characterized by a dramatic increase in smooth muscle and airway inflammation. Although glucocorticoids are the mainstay of treatment in asthma, they are unable to fully control the disease in individuals with severe asthma. We found that airway smooth muscle cells (ASMCs) from individuals with severe asthma showed elevated activities of the ERK1/ERK2 and p38 MAPK pathways despite treatment with oral and inhaled glucocorticoids, which increased the expression of DUSP1, a phosphatase shown to limit p38 MAPK activity. In ex vivo ASMCs, TNF-α but not IL-17A induced expression of the neutrophil chemoattractant CXCL8. Moreover, TNF-α led to up-regulation of the ERK1/ERK2 and p38 MAPKs pathways, with only the latter being sensitive to pretreatment with the glucocorticoid dexamethasone. In contrast to epithelial and endothelial cells, TNF-α-stimulated CXCL8 synthesis was dependent on ERK1/ERK2 but not on p38 MAPK. Moreover, suppressing ERK1/ERK2 activation prevented neutrophil recruitment by ASMCs, whereas suppressing p38 MAPK activity had no impact. Taken together, these results highlight the ERK1/ERK2 MAPK cascade as a novel and attractive target in severe asthma because the activation of this pathway is insensitive to the action of glucocorticoids and is involved in neutrophil recruitment, contributing the to inflammation seen in the disease.


Subject(s)
Asthma/metabolism , Interleukin-8/biosynthesis , MAP Kinase Signaling System , Neutrophils/metabolism , Adult , Aged , Aged, 80 and over , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Asthma/pathology , Cells, Cultured , Dual Specificity Phosphatase 1/biosynthesis , Female , Glucocorticoids/therapeutic use , Humans , Male , Middle Aged , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Severity of Illness Index , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , Young Adult
16.
Am J Physiol Lung Cell Mol Physiol ; 300(6): L958-66, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21460123

ABSTRACT

IL-13 is an important mediator of allergen-induced airway hyperresponsiveness. This Th2 cytokine, produced by activated T cells, mast cells, and basophils, has been described to mediate a part of its effects independently of inflammation through a direct modulation of the airway smooth muscle (ASM). Previous studies demonstrated that IL-13 induces hyperresponsiveness in vivo and enhances calcium signaling in response to contractile agonists in vitro. We hypothesized that IL-13 drives human ASM cells (ASMC) to a procontractile phenotype. We evaluated ASM phenotype through the ability of the cell to proliferate, to contract, and to express contractile protein in response to IL-13. We found that IL-13 inhibits human ASMC proliferation (expression of Ki67 and bromodeoxyuridine incorporation) in response to serum, increasing the number of cells in G0/G1 phase and decreasing the number of cells in G2/M phases of the cell cycle. IL-13-induced inhibition of proliferation was not dependent on signal transducer and activator of transcription-6 but was IL-13Rα2 receptor dependent and associated with a decrease of Kruppel-like factor 5 expression. In parallel, IL-13 increased calcium signaling and the stiffening of human ASMC in response to 1 µM histamine, whereas the stiffening response to 30 mM KCl was unchanged. However, Western blot analysis showed unchanged levels of calponin, smooth muscle α-actin, vinculin, and myosin. We conclude that IL-13 inhibits proliferation via the IL-13Rα2 receptor and induces hypercontractility of human ASMC without change of the phenotypic markers of contractility.


Subject(s)
Bronchi/drug effects , Cell Proliferation/drug effects , Interleukin-13/pharmacology , Muscle Contraction/drug effects , Myocytes, Smooth Muscle/drug effects , Respiratory System/drug effects , Blotting, Western , Bronchi/cytology , Bronchi/metabolism , Calcium Signaling/drug effects , Cell Cycle/drug effects , Cells, Cultured , Contractile Proteins/metabolism , Histamine/pharmacology , Histamine Agonists/pharmacology , Humans , Interleukin-13 Receptor alpha1 Subunit/genetics , Interleukin-13 Receptor alpha1 Subunit/metabolism , Microscopy, Atomic Force , Phenotype , RNA, Messenger/genetics , Respiratory System/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction
17.
Eur J Immunol ; 40(6): 1590-602, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20373517

ABSTRACT

EGF receptor (EGFR) is involved in cell differentiation and proliferation in airways and may trigger cytokine production by T cells. We hypothesized that EGFR inhibition at the time of allergic sensitization may affect subsequent immune reactions. Brown Norway rats were sensitized with OVA, received the EGFR tyrosine kinase inhibitor, AG1478 from days 0 to 7 and OVA challenge on day 14. OVA-specific IgE in serum and cytokines and chemokines in BAL were measured 24 h after challenge. To evaluate effects on airway hyperresponsiveness (AHR), rats were sensitized, treated with AG1478, intranasally challenged, and then AHR was assessed. Furthermore chemotactic activity of BALF for CD4(+) T cells was examined. The eosinophils, neutrophils and lymphocytes in BAL were increased by OVA and only the lymphocytes were reduced by AG1478. OVA significantly enhanced IL-6 concentration in BAL, which was inhibited by AG1478. However AHR, OVA-specific IgE and IL-4 mRNA expression in CD4(+) T cells were not affected by AG1478. BALF from OVA-sensitized/challenged rats induced CD4(+) T-cell migration, which was inhibited by both AG1478 treatment in vivo and neutralization of IL-6 in vitro. EGFR activation during sensitization may affect the subsequent influx of CD4(+) T cells to airways, mainly mediated through IL-6.


Subject(s)
Asthma/immunology , ErbB Receptors/immunology , Interleukin-6/immunology , T-Lymphocytes/immunology , Allergens/immunology , Animals , Asthma/metabolism , Blotting, Western , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cell Separation , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Cytokines/biosynthesis , Disease Models, Animal , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Flow Cytometry , Lung/drug effects , Lung/immunology , Ovalbumin/immunology , Quinazolines , Rats , Rats, Inbred BN , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , Tyrphostins/pharmacology
18.
J Allergy Clin Immunol ; 125(5): 1037-1045.e3, 2010 May.
Article in English | MEDLINE | ID: mdl-20451038

ABSTRACT

BACKGROUND: The origin of the excess airway smooth muscle in asthma and when in the course of the disease it is acquired are uncertain. OBJECTIVES: We examined the relative sensitivities of 2 markers of proliferation, proliferating cell nuclear antigen (PCNA) and Ki 67, in airway smooth muscle in vivo and in vitro. We then studied whether muscle remodeling is a dynamic process in asthma by quantifying proliferation rate and area. Finally we examined heparin-binding epidermal growth factor as a biomarker of remodeling. METHODS: We obtained bronchoscopic biopsies from subjects with moderate or severe asthma and healthy controls (n = 9/group). For in vitro studies, airway smooth muscle cells were cultured from tracheas of transplant donors. The proliferation rate was quantified from PCNA and Ki 67, co-localized to smooth muscle-specific alpha-actin cells in vivo. Muscle area was assessed morphometrically. We examined the expression of heparin-binding epidermal growth factor on tissues by in situ hybridization and by immunohistochemistry and in cells in culture by RT-PCR. RESULTS: Proliferating cell nuclear antigen and Ki 67 were highly correlated, but PCNA was a significantly more sensitive marker both in vivo and in vitro. Muscle area was 3.4-fold greater and the fraction of PCNA(+) nuclei in muscle was 5-fold greater in severe asthma than in healthy subjects. Heparin-binding epidermal growth factor was upregulated in proliferating muscle cells in culture and in airway smooth muscle in severe asthmatic tissues. CONCLUSION: Proliferating cell nuclear antigen is a highly sensitive marker of proliferation and heparin-binding epidermal growth factor is a potential biomarker during active remodeling of ASM in severe asthma.


Subject(s)
Airway Remodeling/physiology , Asthma/physiopathology , Muscle, Smooth , Severity of Illness Index , Adult , Asthma/pathology , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Ki-67 Antigen/metabolism , Male , Middle Aged , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Polymerase Chain Reaction , Proliferating Cell Nuclear Antigen/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 299(6): L808-15, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20889674

ABSTRACT

Airway epithelial cells release proinflammatory mediators that may contribute to airway remodeling and leukocyte recruitment. We explored the hypothesis that leukotriene D4 (LTD4) may trigger the release of proremodeling factors through activation of the EGF receptor (EGFR). We particularly focused on the effects of LTD4 on release of heparin-binding EGF-like factor (HB-EGF) and IL-8 (CXCL8), a potent neutrophil chemoattractant that may be released downstream of EGFR activation. To address this hypothesis, both primary (NHBE) and transformed bronchial human epithelial cells (BEAS-2B) were grown on an air-liquid interface and stimulated with LTD4. HB-EGF and CXCL8 were evaluated by ELISA in cell culture supernatants. To explore the EGFR signaling pathway, we used a broad-spectrum matrix metalloproteinase (MMP) inhibitor, GM-6001, two selective EGFR tyrosine kinase inhibitors, AG-1478 and PD-153035, an HB-EGF neutralizing antibody, and a specific small interfering RNA (siRNA) against the EGFR. Expression of the CysLT1 cysteinyl leukotriene receptor was demonstrated by RT-PCR and immunocytochemistry in both BEAS-2B and NHBE cells. Four hours after stimulation with LTD4, HB-EGF and CXCL8 were significantly increased in cell culture supernatant. GM-6001 and montelukast, a specific CysLT1 receptor antagonist, blocked the LTD4-induced increase in HB-EGF. All inhibitors/antagonists decreased LTD4-induced CXCL8 release. siRNA against EGFR abrogated CXCL8 release following stimulation with LTD4 and exogenous HB-EGF. These findings suggest LTD4 induced EGFR transactivation through the release of HB-EGF in human bronchial epithelial cells with downstream release of CXCL8. These effects may contribute to epithelial-mediated airway remodeling in asthma and other conditions associated with cysteinyl leukotriene release.


Subject(s)
Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-8/metabolism , Leukotriene D4/pharmacology , Respiratory Mucosa/cytology , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells/cytology , ErbB Receptors/genetics , Heparin-binding EGF-like Growth Factor , Humans , Leukotriene D4/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
20.
Respir Res ; 11: 139, 2010 Oct 07.
Article in English | MEDLINE | ID: mdl-20929541

ABSTRACT

BACKGROUND: Human hemokinin-1 (hHK-1) and endokinins are peptides of the tachykinin family encoded by the TAC4 gene. TAC4 and hHK-1 expression as well as effects of hHK-1 in the lung and airways remain however unknown and were explored in this study. METHODS: RT-PCR analysis was performed on human bronchi to assess expression of tachykinin and tachykinin receptors genes. Enzyme immunoassay was used to quantify hHK-1, and effects of hHK-1 and endokinins on contraction of human and guinea pig airways were then evaluated, as well as the role of hHK-1 on cytokines production by human lung parenchyma or bronchi explants and by lung macrophages. RESULTS: In human bronchi, expression of the genes that encode for hHK-1, tachykinin NK1-and NK2-receptors was demonstrated. hHK-1 protein was found in supernatants from explants of human bronchi, lung parenchyma and lung macrophages. Exogenous hHK-1 caused a contractile response in human bronchi mainly through the activation of NK2-receptors, which blockade unmasked a NK1-receptor involvement, subject to a rapid desensitization. In the guinea pig trachea, hHK-1 caused a concentration-dependant contraction mainly mediated through the activation of NK1-receptors. Endokinin A/B exerted similar effects to hHK-1 on both human bronchi and guinea pig trachea, whereas endokinins C and D were inactive. hHK-1 had no impact on the production of cytokines by explants of human bronchi or lung parenchyma, or by human lung macrophages. CONCLUSIONS: We demonstrate endogenous expression of TAC4 in human bronchi, the encoded peptide hHK-1 being expressed and involved in contraction of human and guinea pig airways.


Subject(s)
Bronchi/physiology , Bronchoconstriction/physiology , Gene Expression Regulation , Tachykinins/biosynthesis , Animals , Female , Guinea Pigs , Humans , Male , Species Specificity , Tachykinins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL