Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 414
Filter
Add more filters

Publication year range
1.
J Biol Chem ; 300(4): 107127, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38432633

ABSTRACT

Regulators of G protein signaling (RGS) proteins constrain G protein-coupled receptor (GPCR)-mediated and other responses throughout the body primarily, but not exclusively, through their GTPase-activating protein activity. Asthma is a highly prevalent condition characterized by airway hyper-responsiveness (AHR) to environmental stimuli resulting in part from amplified GPCR-mediated airway smooth muscle contraction. Rgs2 or Rgs5 gene deletion in mice enhances AHR and airway smooth muscle contraction, whereas RGS4 KO mice unexpectedly have decreased AHR because of increased production of the bronchodilator prostaglandin E2 (PGE2) by lung epithelial cells. Here, we found that knockin mice harboring Rgs4 alleles encoding a point mutation (N128A) that sharply curtails RGS4 GTPase-activating protein activity had increased AHR, reduced airway PGE2 levels, and augmented GPCR-induced bronchoconstriction compared with either RGS4 KO mice or WT controls. RGS4 interacted with the p85α subunit of PI3K and inhibited PI3K-dependent PGE2 secretion elicited by transforming growth factor beta in airway epithelial cells. Together, these findings suggest that RGS4 affects asthma severity in part by regulating the airway inflammatory milieu in a G protein-independent manner.


Subject(s)
Asthma , RGS Proteins , Animals , Humans , Mice , Asthma/metabolism , Asthma/genetics , Asthma/pathology , Bronchoconstriction/genetics , Dinoprostone/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Mice, Knockout , Phosphatidylinositol 3-Kinases/metabolism , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , RGS Proteins/metabolism , RGS Proteins/genetics , Cell Line
2.
J Allergy Clin Immunol ; 148(3): 843-857.e6, 2021 09.
Article in English | MEDLINE | ID: mdl-33684437

ABSTRACT

BACKGROUND: Prenatal exposure to infections can modify immune development. These environmental disturbances during early life potentially alter the incidence of inflammatory disorders as well as priming of immune responses. Infection with the helminth Schistosoma mansoni is widely studied for its ability to alter immune responsiveness and is associated with variations in coinfection, allergy, and vaccine efficacy in endemic populations. OBJECTIVE: Exposure to maternal schistosomiasis during early life, even without transmission of infection, can result in priming effects on offspring immune responses to bystander antigenic challenges as related to allergic responsiveness and vaccination, with this article seeking to further clarify the effects and underlying immunologic imprinting. METHODS: Here, we have combined a model of chronic maternal schistosomiasis infection with a thorough analysis of subsequent offspring immune responses to allergy and vaccination models, including viral challenge and steady-state changes to immune cell compartments. RESULTS: We have demonstrated that maternal schistosomiasis alters CD4+ responses during allergic sensitization and challenge in a skewed IL-4/B-cell-dominant response to antigenic challenge associated with limited inflammatory response. Beyond that, we have uncovered previously unidentified alterations to CD8+ T-cell responses during immunization that are dependent on vaccine formulation and have functional impact on the efficacy of vaccination against viral infection in a murine hepatitis B virus model. CONCLUSION: In addition to steady-state modifications to CD4+ T-cell polarization and B-cell priming, we have traced these modified CD8+ responses to an altered dendritic cell phenotype sustained into adulthood, providing evidence for complex priming effects imparted by infection via fetomaternal cross talk.


Subject(s)
Prenatal Exposure Delayed Effects/immunology , Respiratory Hypersensitivity/immunology , Schistosomiasis/immunology , Allergens/immunology , Animals , B-Lymphocytes/immunology , Cells, Cultured , Dendritic Cells/immunology , Female , Fetus/immunology , Gene Expression Profiling , Immunization , Lung/immunology , Lymph Nodes/immunology , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Ovalbumin/immunology , Pregnancy , Respiratory Hypersensitivity/genetics , Schistosoma mansoni , Spleen/immunology , T-Lymphocytes/immunology
3.
J Cell Mol Med ; 25(14): 6573-6583, 2021 07.
Article in English | MEDLINE | ID: mdl-34050597

ABSTRACT

Asthma is a serious public health problem worldwide, without effective therapeutic methods. Our previous study indicated that glucocorticoid-induced transcript 1 gene (GLCCI1) knockout reduces the sensitivity to glucocorticoid in asthmatic mouse. Here, we explored the role and action mechanism of GLCCI1 in asthma development. In ovalbumin-sensitized mice, airway resistance and tissue damage increased, the production of inflammatory cytokines were up-regulated, GLCCI1 expression was reduced and autophagy was activated. Increasing of GLCCI1 inhibited human and mouse airway epithelial cell (AEC) autophagy, while decreasing of GLCCI1 promoted autophagy. Furthermore, we found that GLCCI1 bound with WD repeat domain 45B (WDR45B) and inhibited its expression. Increasing of WDR45B partly reversed the inhibition of GLCCI1 to autophagy-related proteins expression and autophagosome formation in vitro. Increasing of WDR45B in vivo reversed the improvement of GLCCI1 on airway remodelling in asthma and the inhibition to autophagy level in lung tissues. Overall, our data showed that GLCCI1 improved airway remodelling in ovalbumin-sensitized mice through inhibiting autophagy via combination with WDR45B and inhibiting its expression. Our results proved a new idea for asthma treatment.


Subject(s)
Asthma/genetics , Collagen/metabolism , Receptors, Glucocorticoid/genetics , Respiratory Hypersensitivity/genetics , Administration, Inhalation , Airway Remodeling/genetics , Animals , Asthma/pathology , Asthma/therapy , Autophagy/genetics , Autophagy-Related Proteins/genetics , Disease Models, Animal , Humans , Lung/metabolism , Mice , Protein Binding/genetics , Respiratory Hypersensitivity/pathology , WD40 Repeats/genetics
4.
Clin Exp Allergy ; 51(4): 564-573, 2021 04.
Article in English | MEDLINE | ID: mdl-33471392

ABSTRACT

BACKGROUND: In asthma, IL-6 is a potential cause of enhanced inflammation, tissue damage and airway dysfunction. IL-6 signalling is regulated by its receptor, which is composed of two proteins, IL-6R and GP130. In addition to their membrane form, these two proteins may be found as extracellular soluble forms. The interaction of IL-6 with soluble IL-6R (sIL-6R) can trigger IL-6 trans-signalling in cells lacking IL-6R. Conversely, the soluble form of GP130 (sGP130) competes with its membrane form to inhibit IL-6 trans-signalling. OBJECTIVES: We aimed to analyse IL-6 trans-signalling proteins in the airways of subjects after an allergen challenge. METHODS: We used a model of segmental bronchoprovocation with an allergen (SBP-Ag) in human subjects with allergy. Before and 48 h after SBP-Ag, bronchoalveolar lavages (BALs) allowed for the analysis of proteins in BAL fluids (BALFs) by ELISA, and membrane proteins on the surface of BAL cells by flow cytometry. In addition, we performed RNA sequencing (RNA-seq) and used proteomic data to further inform on the expression of the IL-6R subunits by eosinophils, bronchial epithelial cells and lung fibroblasts. Finally, we measured the effect of IL-6 trans-signalling on bronchial fibroblasts, in vitro. RESULTS: IL-6, sIL-6R, sGP130 and the molar ratio of sIL-6R/sGP130 increased in the airways after SBP-Ag, suggesting the potential for enhanced IL-6 trans-signalling activity. BAL lymphocytes, monocytes and eosinophils displayed IL-6R on their surface and were all possible providers of sIL-6R, whereas GP130 was highly expressed in bronchial epithelial cells and lung fibroblasts. Finally, bronchial fibroblasts activated by IL-6 trans-signalling produced enhanced amounts of the chemokine, MCP-1 (CCL2). CONCLUSION AND CLINICAL RELEVANCE: After a bronchial allergen challenge, we found augmentation of the elements of IL-6 trans-signalling. Allergen-induced IL-6 trans-signalling activity can activate fibroblasts to produce chemokines that can further enhance inflammation and lung dysfunction.


Subject(s)
Asthma/metabolism , Cytokine Receptor gp130/metabolism , Interleukin-6/metabolism , Receptors, Interleukin-6/metabolism , Allergens , Ambrosia , Animals , Asthma/genetics , Bronchial Provocation Tests , Bronchoalveolar Lavage Fluid/chemistry , Chemokine CCL2/metabolism , Cytokine Receptor gp130/genetics , Dander , Female , Humans , Interleukin-6/genetics , Male , Pyroglyphidae , RNA-Seq , Receptors, Interleukin-6/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/metabolism , Young Adult
5.
Int J Mol Sci ; 22(15)2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34361007

ABSTRACT

The immune system defends the body against certain tumor cells and against foreign agents such as fungi, parasites, bacteria, and viruses. One of its main roles is to distinguish endogenous components from non-self-components. An unproperly functioning immune system is prone to primary immune deficiencies caused by either primary immune deficiencies such as genetic defects or secondary immune deficiencies such as physical, chemical, and in some instances, psychological stressors. In the manuscript, we will provide a brief overview of the immune system and immunotoxicology. We will also describe the biochemical mechanisms of immunotoxicants and how to evaluate immunotoxicity.


Subject(s)
Allergens/toxicity , Environmental Illness/immunology , Food Hypersensitivity/immunology , Respiratory Hypersensitivity/immunology , Allergens/immunology , Animals , Environmental Illness/genetics , Food Hypersensitivity/genetics , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Respiratory Hypersensitivity/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/immunology
6.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681784

ABSTRACT

Mitochondria regulate a myriad of cellular functions. Dysregulation of mitochondrial control within airway epithelial cells has been implicated in the pro-inflammatory response to allergens in asthma patients. Because of their multifaceted nature, mitochondrial structure must be tightly regulated through fission and fusion. Dynamin Related Protein 1 (DRP1) is a key driver of mitochondrial fission. During allergic asthma, airway epithelial mitochondria appear smaller and structurally altered. The role of DRP1-mediated mitochondrial fission, however, has not been fully elucidated in epithelial response to allergens. We used a Human Bronchial Epithelial Cell line (HBECs), primary Mouse Tracheal Epithelial Cells (MTECs), and conditional DRP1 ablation in lung epithelial cells to investigate the impact of mitochondrial fission on the pro-inflammatory response to house dust mite (HDM) in vitro and in vivo. Our data suggest that, following HDM challenge, mitochondrial fission is rapidly upregulated in airway epithelial cells and precedes production of pro-inflammatory cytokines and chemokines. Further, deletion of Drp1 in lung epithelial cells leads to decreased fission and enhanced pro-inflammatory signaling in response to HDM in vitro, as well as enhanced airway hyper-responsiveness (AHR), inflammation, differential mucin transcription, and epithelial cell death in vivo. Mitochondrial fission, therefore, regulates the lung epithelial pro-inflammatory response to HDM.


Subject(s)
Allergens/pharmacology , Dynamins/physiology , Mitochondrial Dynamics/genetics , Respiratory Hypersensitivity/genetics , Respiratory Mucosa/drug effects , Animals , Bronchi/drug effects , Bronchi/physiology , Cells, Cultured , Dynamins/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Mice , Mice, Transgenic , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism
7.
Lung ; 198(1): 105-112, 2020 02.
Article in English | MEDLINE | ID: mdl-31820077

ABSTRACT

BACKGROUND: Airway epithelial cells secrete Interleukin-33 in response to the different allergens. Several single nucleotide polymorphisms (SNP) of this cytokine have been reported to be involved in the development of asthma. We conducted this study to evaluate the impact of the two most common SNPs of the IL-33 gene (rs1342326 and rs3939286) and environmental factors on the susceptibility to asthma in the Iranian population. SUBJECTS AND METHODS: In this study, we enrolled 126 asthmatics patients and 300 age, sex-matched controls. Genotyping was performed by real-time PCR using the TaqMan SNP genotyping assay. Moreover, total serum IgE level, eosinophil count, and skin prick test were accomplished and complete history was taken from all the participants. RESULTS: The frequencies of mutant genotypes in both SNPs were significantly higher in asthmatics than controls. C/C genotype of rs1342326 [OR (95% CI) 2.50 (1.33-4.69)] and A/A genotype of rs3939286 [OR (95% CI) 2.18 (1.05-4.52)] were associated with higher risk of asthma development. While A/C+C/C genotype of rs1342326 was more prevalent in mild asthma [OR (95% CI) 2.36 (1.14-4.89)], G/A+A/A genotype of rs3939286 was associated with increased risk of moderate and severe asthma [OR (95% CI) 2.53 (1.30-4.94)]. CONCLUSION: This study revealed that both IL-33 SNPs were associated with an increased risk of asthma. The rs1342326 was associated with atopic, mild and adult-onset asthma and a higher level of eosinophils in peripheral blood. However, rs3939286 was more frequent in moderate and severe asthma. Moreover, rs3939286 was associated with non-atopic and childhood-onset asthma.


Subject(s)
Asthma/genetics , Eosinophilia/genetics , Gene-Environment Interaction , Interleukin-33/genetics , Adult , Age of Onset , Asthma/epidemiology , Asthma/immunology , Asthma/physiopathology , Case-Control Studies , Eosinophilia/epidemiology , Eosinophilia/immunology , Female , Forced Expiratory Volume , Genetic Predisposition to Disease , Genotype , Humans , Immunoglobulin E/immunology , Iran/epidemiology , Male , Middle Aged , Polymorphism, Single Nucleotide , Respiratory Hypersensitivity/epidemiology , Respiratory Hypersensitivity/genetics , Risk Factors , Severity of Illness Index , Skin Tests , Vital Capacity
8.
Am J Respir Cell Mol Biol ; 60(4): 399-412, 2019 04.
Article in English | MEDLINE | ID: mdl-30335467

ABSTRACT

The asthma candidate gene inositol polyphosphate 4-phosphatase type I A (INPP4A) is a lipid phosphatase that negatively regulates the PI3K/Akt pathway. Destabilizing genetic variants of INPP4A increase the risk of asthma, and lung-specific INPP4A knockdown induces asthma-like features. INPP4A is known to localize intracellularly, and its extracellular presence has not been reported yet. Here we show for the first time that INPP4A is secreted by airway epithelial cells and that extracellular INPP4A critically inhibits airway inflammation and remodeling. INPP4A was present in blood and BAL fluid, and this extracellular INPP4A was reduced in patients with asthma and mice with allergic airway inflammation. In both naive mice and mice with allergic airway inflammation, antibody-mediated neutralization of extracellular INPP4A potentiated PI3K/Akt signaling and induced airway hyperresponsiveness, with prominent airway remodeling, subepithelial fibroblast proliferation, and collagen deposition. The link between extracellular INPP4A and fibroblasts was investigated in vitro. Cultured airway epithelial cells secreted enzymatically active INPP4A in extracellular vesicles and in a free form. Extracellular vesicle-mediated transfer of labeled INPP4A, from epithelial cells to fibroblasts, was observed. Inhibition of such transfer by anti-INPP4A antibody increased fibroblast proliferation. We propose that secretory INPP4A is a novel "paracrine" layer of the intricate regulation of lung homeostasis, by which airway epithelium dampens PI3K/Akt signaling in inflammatory cells or local fibroblasts, thereby limiting inflammation and remodeling.


Subject(s)
Airway Remodeling/physiology , Asthma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Respiratory Hypersensitivity/pathology , Airway Remodeling/genetics , Animals , Asthma/blood , Asthma/genetics , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Fibroblasts/metabolism , Humans , Lung/pathology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Phosphoric Monoester Hydrolases/blood , Phosphoric Monoester Hydrolases/genetics , Respiratory Hypersensitivity/genetics , Signal Transduction/genetics
9.
Biochem Biophys Res Commun ; 516(3): 739-746, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31255288

ABSTRACT

Orosomucoid-like protein 3 (ORMDL3) is a common mutation in many asthma patients and its effects on the specific pathogenesis of asthma are still unclear. Therefore, in this study, we used a mouse that specifically knockout the mouse ORDML3 gene to further study the mechanism. We used ovalbumin (OVA) to induce asthma in wild-type mice and ORMDL3 knockout mice. Lung ventilation resistance, airway inflammation, mucus hypersecretion, collagen deposition, the levels of inflammatory factors and the expression of ORDML3 and JNK1/2-MMP-9 pathway were detected. The results showed that ORMDL3 gene was highly expressed in clinical asthmatic children and mouse asthma model. Knocking down the ORMDL3 gene in the lung tissue of asthmatic mice can reduce airway hyperresponsiveness, airway inflammation, mucus secretion, and collagen deposition around the airway. After knocking down the lung tissue of mice, the IL-4, IL-5 and IL-13 concentrations in broncho alveolar lavage fluid of asthmatic mice were significantly decreased, and the activation of JNK1/2-MMP-9 pathway was inhibited in mouse lung tissue. Collectively, our results demonstrate that the ORMDL3 gene may aggravate asthma symptoms by activating the JNK1/2-MMP-9 pathway, which indicates that the ORMDL3 gene may be the key molecule for the next step of asthma targeted therapy.


Subject(s)
Asthma/genetics , Matrix Metalloproteinase 9/metabolism , Membrane Proteins/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Respiratory Hypersensitivity/genetics , Airway Remodeling/drug effects , Animals , Asthma/chemically induced , Asthma/physiopathology , Asthma/prevention & control , Bronchoalveolar Lavage Fluid/chemistry , Child , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Interleukin-13/genetics , Interleukin-13/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Interleukin-5/genetics , Interleukin-5/metabolism , Lung/drug effects , Lung/metabolism , Lung/physiopathology , Matrix Metalloproteinase 9/genetics , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/genetics , Mucus/chemistry , Mucus/metabolism , Ovalbumin/administration & dosage , Respiratory Function Tests , Respiratory Hypersensitivity/chemically induced , Respiratory Hypersensitivity/physiopathology , Respiratory Hypersensitivity/prevention & control , Signal Transduction
10.
Toxicol Appl Pharmacol ; 368: 55-62, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30682383

ABSTRACT

Quantum dot nanoparticles (QDs) are engineered nanomaterials (ENMs) that have utility in many industries due to unique optical properties not available in small molecules or bulk materials. QD-induced acute lung inflammation and toxicity in rodent models raise concerns about potential human health risks. Recent studies have also shown that some ENMs can exacerbate allergic airway disease (AAD). In this study, C57BL/6J and A/J mice were exposed to saline, house dust mite (HDM), or a combination of HDM and QDs on day 1 of the sensitization protocol. Mice were then challenged on days 8, 9 and 10 with HDM or saline only. Significant differences in cellular and molecular markers of AAD induced by both HDM and HDM + QD were observed between C57BL/6J and A/J mice. Among A/J mice, HDM + QD co-exposure, but not HDM exposure alone, significantly increased levels of bronchoalveolar lavage fluid (BALF). IL-33 compared to saline controls. BALF total protein levels in both mouse strains were also only significantly increased by HDM + QD co-exposure. In addition, A/J mice had significantly more lung type 2 innate lymphoid cells (ILC2s) cells than C57BL/6J mice. A/J lung ILC2s were inversely correlated with lung glutathione and MHC-IIhigh resident macrophages, and positively correlated with MHC-IIlow resident macrophages. The results from this study suggest that 1) QDs influence HDM-induced AAD by potentiating and/or enhancing select cytokine production; 2) that genetic background modulates the impact of QDs on HDM sensitization; and 3) that potential ILC2 contributions to HDM induced AAD are also likely to be modulated by genetic background.


Subject(s)
Antigens, Dermatophagoides/immunology , Insect Proteins/immunology , Lung/drug effects , Pyroglyphidae/immunology , Quantum Dots/toxicity , Respiratory Hypersensitivity/chemically induced , Animals , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Genotype , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lung/immunology , Lung/metabolism , Lung/physiopathology , Male , Mice, Inbred C57BL , Phenotype , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/physiopathology , Risk Factors , Species Specificity
11.
Int Arch Allergy Immunol ; 180(3): 182-194, 2019.
Article in English | MEDLINE | ID: mdl-31412349

ABSTRACT

Asthma is a complex chronic disease and the pathogenesis is still not entirely clear. In this study, we aimed to clarify the role and mechanism of miR-29b in the development of asthma. We observed that miR-29b levels were decreased in the lung and spleen of OVA-induced asthmatic mice. Reverse transcription-quantitative polymerase chain reaction and flow cytometry demonstrated that the inducible co-stimulator (ICOS) expression at mRNA and protein levels was elevated in the lung of asthmatic mice, and miR-29b expression in the lung of asthmatic mice was negatively associated with ICOS mRNA levels by Pearson Correlation analysis. Additional, flow cytometry showed that the percentage of CD4+ICOS+ T cells in the lung and spleen was regulated by miR-29b, and dual luciferase reporter assay confirmed ICOS was a target gene of miR-29b. Furthermore, miR-29b overexpression in asthmatic mice was induced with miR-29b agomir by intranasal administration; miR-29b alleviated total inflammatory cell infiltration and CCL24 levels, decreased IL-5 levels in bronchoalveolar lavage fluid and serum, and upregulated IFN-γ expression in serum. This study demonstrates that miR-29b targets ICOS, thereby reverses the imbalance of T helper 1 cells (Th1)/Th2 responses and decreases eosinophils recruitment in the airway, which are key features of allergic airway inflammation. Therefore, miR-29b might be an attractive candidate target for asthma treatment.


Subject(s)
Asthma/genetics , Eosinophils/immunology , Lung/immunology , MicroRNAs/genetics , Respiratory Hypersensitivity/genetics , Th1 Cells/immunology , Th2 Cells/immunology , Allergens/immunology , Animals , Cell Movement , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , RNA, Small Interfering/genetics , Th1-Th2 Balance
12.
J Pathol ; 244(4): 485-498, 2018 04.
Article in English | MEDLINE | ID: mdl-29359814

ABSTRACT

Pulmonary vascular remodeling is the main pathological hallmark of pulmonary hypertension disease. We undertook a comprehensive and multilevel approach to investigate the origin of smooth muscle actin-expressing cells in remodeled vessels. Transgenic mice that allow for specific, inducible, and permanent labeling of endothelial (Cdh5-tdTomato), smooth muscle (Acta2-, Myh11-tdTomato), pericyte (Cspg4-tdTomato), and fibroblast (Pdgfra-tdTomato) lineages were used to delineate the cellular origins of pulmonary vascular remodeling. Mapping the fate of major lung resident cell types revealed smooth muscle cells (SMCs) as the predominant source of cells that populate remodeled pulmonary vessels in chronic hypoxia and allergen-induced murine models. Combining in vivo cell type-specific, time-controlled labeling of proliferating cells with a pulmonary artery phenotypic explant assay, we identified proliferation of SMCs as an underlying remodeling pathomechanism. Multicolor immunofluorescence analysis showed a preserved pattern of cell type marker localization in murine and human pulmonary arteries, in both donors and idiopathic pulmonary arterial hypertension (IPAH) patients. Whilst neural glial antigen 2 (chondroitin sulfate proteoglycan 4) labeled mostly vascular supportive cells with partial overlap with SMC markers, PDGFRα-expressing cells were observed in the perivascular compartment. The luminal vessel side was lined by a single cell layer expressing endothelial markers followed by an adjacent and distinct layer defined by SMC marker expression and pronounced thickening in remodeled vessels. Quantitative flow cytometric analysis of single cell digests of diverse pulmonary artery layers showed the preserved separation into two discrete cell populations expressing either endothelial cell (EC) or SMC markers in human remodeled vessels. Additionally, we found no evidence of overlap between EC and SMC ultrastructural characteristics using electron microscopy in either donor or IPAH arteries. Lineage-specific marker expression profiles are retained during pulmonary vascular remodeling without any indication of cell type conversion. The expansion of resident SMCs is the major underlying and evolutionarily conserved paradigm of pulmonary vascular disease pathogenesis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Subject(s)
Cell Lineage , Genes, Reporter , Hypoxia/pathology , Lung/blood supply , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Respiratory Hypersensitivity/pathology , Vascular Remodeling , Actins/genetics , Actins/metabolism , Animals , Antigens/genetics , Antigens/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Chronic Disease , Disease Models, Animal , Familial Primary Pulmonary Hypertension/metabolism , Familial Primary Pulmonary Hypertension/pathology , Familial Primary Pulmonary Hypertension/physiopathology , Fluorescent Antibody Technique , Humans , Hypoxia/genetics , Hypoxia/metabolism , Hypoxia/physiopathology , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/metabolism , Phenotype , Proteoglycans/genetics , Proteoglycans/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/physiopathology , Red Fluorescent Protein
13.
Crit Rev Eukaryot Gene Expr ; 28(2): 125-127, 2018.
Article in English | MEDLINE | ID: mdl-30055538

ABSTRACT

MicroRNAs (miRNAs) are short noncoding RNAs that control gene expression posttranscriptionally by directly blocking translation of their target mRNAs or by repressing protein production via mRNA destabilization. Investigations into miRNAs began approximately 12 years ago with their discovery in mammalian cells. Still, the involvement of miRNAs in the development of asthma remains unclear, and this topic needs further research to discover various molecular mechanisms responsible for the pathogenesis of asthma and new therapeutic interventions. So far, various miRNAs have been identified in allergic airway disease along with their targets. Our present mini-review highlights the latest information involving the role of miRNAs in asthma.


Subject(s)
Asthma/genetics , MicroRNAs/genetics , Respiratory Hypersensitivity/genetics , Asthma/pathology , Gene Expression Regulation/genetics , Humans , RNA, Messenger/genetics , Respiratory Hypersensitivity/pathology , Respiratory System/pathology
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 934-941, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29305916

ABSTRACT

Asthma is a chronic airway inflammation in which Th2 and Th17 cells play critical roles in its pathogenesis. We have reported that atypical protein kinase (PKC) λ/ι is a new regulator for Th2 differentiation and function. However, the role of PKCλ/ι for Th17 cells remains elusive. In this study, we explored the effect of PKCλ/ι on Th17 cells in the context of ex vivo cell culture systems and an in vivo murine model of allergic airway inflammation with the use of activated T cell-specific conditional PKCλ/ι-deficient mice. Our findings indicate that PKCλ/ι regulates Th17 cells. The secretion of Th17 effector cytokines, including IL-17, IL-21 and IL-22, were inhibited from PKCλ/ι-deficient T cells under non-skewing or Th17-skewing culture conditions. Moreover, the impaired Th17 differentiation and function by the PKCλ/ι-deficiency was associated with the downregulation of Stat3 and Rorγt, key Th17 transcription factors. We developed a model of Th17 and neutrophil-involved allergic airway inflammation by intratracheal inoculation of house dust mites. PKCλ/ι-deficiency significantly inhibited airway inflammations. The infiltrating cells in the lungs and bronchoalveolar lavage fluids were significantly reduced in conditional PKCλ/ι-deficient mice. Th17 effector cytokines were reduced in the bronchoalveolar lavage fluids and lungs at protein and mRNA levels. Thus, PKCλ/ι emerges as a critical regulator of Th17 differentiation and allergic airway hyperresponsiveness.


Subject(s)
Cell Differentiation/genetics , Inflammation , Isoenzymes/physiology , Protein Kinase C/physiology , Pyroglyphidae/immunology , Respiratory Hypersensitivity , Th17 Cells/physiology , Animals , Dermatophagoides pteronyssinus/immunology , Embryo, Mammalian , Female , Inflammation/genetics , Inflammation/immunology , Isoenzymes/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Protein Kinase C/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology
15.
Mamm Genome ; 29(3-4): 281-298, 2018 04.
Article in English | MEDLINE | ID: mdl-29536159

ABSTRACT

The human chromosomal region 17q12-q21 is one of the best replicated genome-wide association study loci for childhood asthma. The associated SNPs span a large genomic interval that includes several protein-coding genes. Here, we tested the hypothesis that the zona pellucida-binding protein 2 (ZPBP2) gene residing in this region contributes to asthma pathogenesis using a mouse model. We tested the lung phenotypes of knock-out (KO) mice that carry a deletion of the Zpbp2 gene. The deletion attenuated airway hypersensitivity (AHR) in female, but not male, mice in the absence of allergic sensitization. Analysis of the lipid profiles of their lungs showed that female, but not male, KO mice had significantly lower levels of sphingosine-1-phosphate (S1P), very long-chain ceramides (VLCCs), and higher levels of long-chain ceramides compared to wild-type controls. Furthermore, in females, lung resistance following methacholine challenge correlated with lung S1P levels (Pearson correlation coefficient 0.57) suggesting a link between reduced AHR in KO females, Zpbp2 deletion, and S1P level regulation. In livers, spleens and blood plasma, however, VLCC, S1P, and sphingosine levels were reduced in both KO females and males. We also find that the Zpbp2 deletion was associated with gain of methylation in the adjacent DNA regions. Thus, we demonstrate that the mouse ortholog of ZPBP2 has a role in controlling AHR in female mice. Our data also suggest that Zpbp2 may act through regulation of ceramide metabolism. These findings highlight the importance of phospholipid metabolism for sexual dimorphism in AHR.


Subject(s)
Lipid Metabolism , Lung/metabolism , Membrane Proteins/genetics , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/pathology , Sex Characteristics , Animals , Asthma/genetics , Asthma/pathology , DNA Methylation/genetics , Disease Models, Animal , Female , Gene Deletion , Gene Expression Regulation , Immunoglobulin E/metabolism , Liver/metabolism , Liver/pathology , Lung/pathology , MAP Kinase Signaling System , Male , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Methacholine Chloride , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Phenotype , Promoter Regions, Genetic , Sphingolipids/metabolism , Transcriptome/genetics
16.
Clin Sci (Lond) ; 132(3): 327-338, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29269381

ABSTRACT

Airway remodelling and allergic inflammation are key features of airway hyperresponsiveness (AHR) in asthma; however, their interrelationships are unclear. The present study investigated the separate and combined effects of increased airway smooth muscle (ASM) layer thickness and allergy on AHR. We integrated a protocol of ovalbumin (OVA)-induced allergy into a non-inflammatory mouse model of ASM remodelling induced by conditional and airway-specific expression of transforming growth factor-α (TGF-α) in early growth response-1 (Egr-1)-deficient transgenic mice, which produced thickening of the ASM layer following ingestion of doxycycline. Mice were sensitised to OVA and assigned to one of four treatment groups: Allergy - normal chow diet and OVA challenge; Remodelling - doxycycline in chow and saline challenge; Allergy and Remodelling - doxycycline in chow and OVA challenge; and Control - normal chow diet and saline challenge. Airway responsiveness to methacholine (MCh) and histology were assessed. Compared with the Control group, airway responsiveness to MCh was increased in the Allergy group, independent of changes in wall structure, whereas airway responsiveness in the Remodelling group was increased independent of exposure to aeroallergen. The combined effects of allergy and remodelling on airway responsiveness were greater than either of them alone. There was a positive relationship between the thickness of the ASM layer with airway responsiveness, which was shifted upward in the presence of allergy. These findings support allergy and airway remodelling as independent causes of variable and excessive airway narrowing.


Subject(s)
Airway Remodeling/immunology , Allergens/immunology , Bronchial Hyperreactivity/immunology , Respiratory Hypersensitivity/immunology , Airway Remodeling/genetics , Animals , Asthma/genetics , Asthma/immunology , Bronchial Hyperreactivity/genetics , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Hypersensitivity/genetics , Mice, Knockout , Muscle, Smooth/immunology , Respiratory Hypersensitivity/genetics
17.
Lung ; 196(3): 297-303, 2018 06.
Article in English | MEDLINE | ID: mdl-29600353

ABSTRACT

PURPOSE: The maintaining of asthma control is difficult due to high variability in response to therapy among patients. Since matrix metalloproteinase 9 (MMP9) is implicated in inflammation and remodeling of asthmatic airways, it could be associated with adequate response to asthma therapy. The aim of this study was to investigate whether variants in 3' end of the MMP9 gene are associated with clinical phenotype and responsiveness to treatment in children with asthma. METHODS: The study included 127 asthmatic children from Slovenia. Variants in the 3' end of the MMP9 gene were analyzed by direct DNA sequencing and the obtained results were correlated with clinical parameters. RESULTS: Two variants were detected, rs13925 and rs20544. For the variant rs20544, statistically significant difference in airway hyperresponsiveness (p = 0.011) and asthma control (p = 0.049) between genotypes was found. Patients with TT genotype had lower airway sensitivity, and after 12 months of treatment showed significant improvement in Asthma Control Test (ACT) scores compared to CC and CT genotype. For the variant rs13925, the association with lung function was observed. The carriers of A allele showed noticeable improvement of lung function after the first 6 months of treatment in comparison to the carriers of G allele (p = 0.046). CONCLUSION: The main finding of our study is the association of MMP9 genotypes rs20544 TT and rs13925 AA and AG with better asthma control, and indirectly better response to treatment. Based on these results, MMP9 deserves further research as a potential predictive biomarker for asthma.


Subject(s)
3' Untranslated Regions/genetics , Asthma/genetics , Matrix Metalloproteinase 9/genetics , Respiratory Hypersensitivity/genetics , Acetates/therapeutic use , Adolescent , Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Asthma/physiopathology , Bronchodilator Agents/therapeutic use , Child , Cyclopropanes , Female , Fluticasone/therapeutic use , Forced Expiratory Volume , Genetic Variation , Humans , Male , Nitric Oxide , Prognosis , Quinolines/therapeutic use , Respiration , Sequence Analysis, DNA , Slovenia , Sulfides , Treatment Outcome , Vital Capacity
18.
Immunol Rev ; 260(1): 235-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24942693

ABSTRACT

The lung, while functioning as a gas exchange organ, encounters a large array of environmental factors, including particulate matter, toxins, reactive oxygen species, chemicals, allergens, and infectious microbes. To rapidly respond to and counteract these elements, a number of innate immune mechanisms have evolved that can lead to lung inflammation and asthma, which is the focus of this review. These innate mechanisms include a role for two incompletely understood cell types, invariant natural killer T (iNKT) cells and innate lymphoid cells (ILCs), which together produce a wide range of cytokines, including interleukin-4 (IL-4), IL-5, IL-13, interferon-γ, IL-17, and IL-22, independently of adaptive immunity and conventional antigens. The specific roles of iNKT cells and ILCs in immunity are still being defined, but both cell types appear to play important roles in the lungs, particularly in asthma. As we gain a better understanding of these innate cell types, we will acquire great insight into the mechanisms by which allergic and non-allergic asthma phenotypes develop.


Subject(s)
Asthma/immunology , Immunity, Innate , Lung/immunology , Adaptive Immunity , Allergens/immunology , Animals , Asthma/genetics , Asthma/metabolism , Asthma/microbiology , Cell Communication , Humans , Lung/metabolism , Lung/microbiology , Lymphocytes/immunology , Lymphocytes/metabolism , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/microbiology
19.
Allergy ; 72(5): 772-782, 2017 May.
Article in English | MEDLINE | ID: mdl-27878828

ABSTRACT

BACKGROUND: Asthma is a Th2 cell-driven inflammatory disease and a major public health concern. The cis-acting element Rad50 hypersensitive site 6 (RHS6) in the Th2 locus control region is essential for regulation of the Th2 cytokine genes; however, its role in allergic airway inflammation and underlying molecular mechanisms of the regulation by RHS6 are poorly understood. OBJECTIVE: We sought to understand the role of RHS6 in the development of allergic airway inflammation and its molecular mechanism for Th2 cytokine expression. METHODS: We used an ovalbumin-induced allergic inflammation model with RHS6-deficient mice to examine the role of RHS6 in this process. To examine molecular mechanism of RHS6 for Th2 cytokine expression, we used DNA affinity chromatography and mass spectrometry, quantitative RT-PCR, ELISA, intracellular cytokine staining, chromatin immunoprecipitation, and co-immunoprecipitation. RESULTS: Deletion of RHS6 caused a dramatic resistance to allergic airway inflammation. RHS6 recruited transcription factors GATA3, SATB1, and IRF4, which play important roles in expression of all three Th2 cytokine genes. RHS6 deficiency caused inhibition of transcription factor-induced Th2 cytokine gene expression. CONCLUSION: RHS6 is a critical regulatory element for allergic airway inflammation and for coordinate regulation of Th2 cytokine genes by recruiting GATA3, SATB1, and IRF4.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Cytokines/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Interferon Regulatory Factors/metabolism , Locus Control Region , Matrix Attachment Region Binding Proteins/metabolism , Th2 Cells/metabolism , Acid Anhydride Hydrolases , Animals , Base Sequence , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , DNA-Binding Proteins , Disease Models, Animal , Genetic Loci , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Protein Binding , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Th2 Cells/immunology
20.
J Immunol ; 194(3): 887-97, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25527785

ABSTRACT

Human studies demonstrated that allergen-specific immunotherapy (IT) represents an effective treatment for allergic diseases. IT involves repeated administration of the sensitizing allergen, indicating a crucial contribution of T cells to its medicinal benefit. However, the underlying mechanisms of IT, especially in a chronic disease, are far from being definitive. In the current study, we sought to elucidate the suppressive mechanisms of IT in a mouse model of chronic allergic asthma. OVA-sensitized mice were challenged with OVA or PBS for 4 wk. After development of chronic airway inflammation, mice received OVA-specific IT or placebo alternately to airway challenge for 3 wk. To analyze the T cell-mediated mechanisms underlying IT in vivo, we elaborated the role of T-bet-expressing Th1 cells, T cell-derived IL-10, and Ag-specific thymic as well as peripherally induced Foxp3(+) regulatory T (Treg) cells. IT ameliorated airway hyperresponsiveness and airway inflammation in a chronic asthma model. Of note, IT even resulted in a regression of structural changes in the airways following chronic inhaled allergen exposure. Concomitantly, IT induced Th1 cells, Foxp3(+), and IL-10-producing Treg cells. Detailed analyses revealed that thymic Treg cells crucially contribute to the effectiveness of IT by promoting IL-10 production in Foxp3-negative T cells. Together with the peripherally induced Ag-specific Foxp3(+) Treg cells, thymic Foxp3(+) Treg cells orchestrate the curative mechanisms of IT. Taken together, we demonstrate that IT is effective in a chronic allergic disease and dependent on IL-10 and thymic as well as peripherally induced Ag-specific Treg cells.


Subject(s)
Asthma/immunology , Asthma/metabolism , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Airway Remodeling , Animals , Asthma/genetics , Asthma/pathology , Asthma/therapy , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Desensitization, Immunologic , Disease Models, Animal , Female , Goblet Cells/pathology , Immunoglobulin E/immunology , Immunomodulation , Immunophenotyping , Interleukin-10/biosynthesis , Lung/immunology , Lung/metabolism , Lung/pathology , Metaplasia , Mice , Mice, Knockout , Ovalbumin/adverse effects , Respiratory Hypersensitivity/genetics , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/metabolism , Respiratory Hypersensitivity/pathology , Respiratory Hypersensitivity/therapy , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL