Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 8721, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457454

ABSTRACT

In asthma, the airway epithelium has an impaired capacity to differentiate and plays a key role in the development of airway inflammation and remodeling through mediator release. The study objective was to investigate the release of (IL)-1 family members from primary airway epithelial-cells during differentiation, and how they affect primary airway fibroblast (PAF)-induced inflammation, extracellular matrix (ECM) production, and collagen I remodeling. The release of IL-1α/ß and IL-33 during airway epithelial differentiation was assessed over 20-days using air-liquid interface cultures. The effect of IL-1 family cytokines on airway fibroblasts grown on collagen-coated well-plates and 3-dimensional collagen gels was assessed by measurement of inflammatory mediators and ECM proteins by ELISA and western blot, as well as collagen fiber formation using non-linear optical microscopy after 24-hours. The production of IL-1α is elevated in undifferentiated asthmatic-PAECs compared to controls. IL-1α/ß induced fibroblast pro-inflammatory responses (CXCL8/IL-8, IL-6, TSLP, GM-CSF) and suppressed ECM-production (collagen, fibronectin, periostin) and the cell's ability to repair and remodel fibrillar collagen I via LOX, LOXL1 and LOXL2 activity, as confirmed by inhibition with ß-aminopropionitrile. These data support a role for epithelial-derived-IL-1 in the dysregulated repair of the asthmatic-EMTU and provides new insights into the contribution of airway fibroblasts in inflammation and airway remodeling in asthma.


Subject(s)
Airway Remodeling/immunology , Asthma/immunology , Collagen/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Respiratory System/cytology , Adolescent , Adult , Case-Control Studies , Cell Differentiation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Male , Middle Aged , Respiratory System/metabolism , Up-Regulation , Young Adult
2.
Respir Res ; 20(1): 150, 2019 Jul 12.
Article in English | MEDLINE | ID: mdl-31299975

ABSTRACT

BACKGROUND: Human rhinovirus (HRV) infections are the primary cause of the common cold and are a major trigger for exacerbations of lower airway diseases, such as asthma and chronic obstructive pulmonary diseases. Although human bronchial epithelial cells (HBE) are the natural host for HRV infections, much of our understanding of how HRV replicates and induces host antiviral responses is based on studies using non-airway cell lines (e.g. HeLa cells). The current study examines the replication cycle of HRV, and host cell responses, in highly differentiated cultures of HBE. METHODS: Highly differentiated cultures of HBE were exposed to initial infectious doses ranging from 104 to 101 50% tissue culture-infective dose (TCID50) of purified HRV-16, and responses were monitored up to 144 h after infection. Viral genomic RNA and negative strand RNA template levels were monitored, along with levels of type I and II interferons and selected antivirals. RESULTS: Regardless of initial infectious dose, relatively constant levels of both genomic and negative strand RNA are generated during replication, with negative strand copy numbers being10,000-fold lower than those of genomic strands. Infections were limited to a small percentage of ciliated cells and did not result in any overt signs of epithelial death. Importantly, regardless of infectious dose, HRV-16 infections were cleared by HBE in the absence of immune cells. Levels of type I and type III interferons (IFNs) varied with initial infectious dose, implying that factors other than levels of double-stranded RNA regulate IFN induction, but the time-course of HRV-16 clearance HBE was the same regardless of levels of IFNs produced. Patterns of antiviral viperin and ISG15 expression suggest they may be generated in an IFN-independent manner during HRV-16 infections. CONCLUSIONS: These data challenge a number of aspects of dogma generated from studies in HeLa cells and emphasize the importance of appropriate cell context when studying HRV infections.


Subject(s)
Cell Differentiation/physiology , Immunity, Innate/physiology , Respiratory Mucosa/physiology , Respiratory Mucosa/virology , Rhinovirus/physiology , Virus Replication/physiology , Cells, Cultured , Humans , Respiratory Mucosa/cytology
3.
Chest ; 148(6): 1508-1516, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26270739

ABSTRACT

In healthy individuals, human rhinovirus (HRV) infections are the major cause of the common cold. These are generally uncomplicated infections except for occasional cases of otitis media or sinusitis. In individuals with asthma, however, HRV infections can have a major impact on disease development and progression. HRV-induced wheezing illnesses in early life are a significant risk factor for subsequent development of asthma, and growing evidence supports a role of recurrent HRV infections in the development and progression of several aspects of airway remodeling in asthma. In addition, HRV infections are one of the most common triggers for acute exacerbations of asthma, which represent a major burden to health-care systems around the world. None of the currently prescribed medications for asthma are effective in preventing or reversing asthma development and airway remodeling or are ideal for treating HRV-induced exacerbations of asthma. Thus, a better understanding of the role of HRV in asthma is important if we are to develop more effective therapies. In the past decade, we have gained new insights into the role of HRV infections in the development and progression of airway remodeling as well as a new appreciation for the proinflammatory and host defense responses to HRV infections that may help to regulate susceptibility to asthma exacerbations. This article reviews the current understanding of the role HRV infections play in the pathogenesis of asthma and identifies possible avenues to new therapeutic strategies for limiting the effects of HRV infections in asthma.


Subject(s)
Asthma , Picornaviridae Infections , Rhinovirus/pathogenicity , Airway Remodeling , Asthma/epidemiology , Asthma/etiology , Asthma/immunology , Asthma/physiopathology , Asthma/virology , Disease Progression , Gene-Environment Interaction , Genetic Predisposition to Disease , Host-Pathogen Interactions/immunology , Humans , Picornaviridae Infections/genetics , Picornaviridae Infections/immunology , Picornaviridae Infections/physiopathology , Risk Factors
4.
Int J Biochem Cell Biol ; 68: 59-69, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26315281

ABSTRACT

The epithelium of asthmatics is characterized by reduced expression of E-cadherin and increased expression of the basal cell markers ck-5 and p63 that is indicative of a relatively undifferentiated repairing epithelium. This phenotype correlates with increased proliferation, compromised wound healing and an enhanced capacity to undergo epithelial-mesenchymal transition (EMT). The transcription factor ß-catenin plays a vital role in epithelial cell differentiation and regeneration, depending on the co-factor recruited. Transcriptional programs driven by the ß-catenin/CBP axis are critical for maintaining an undifferentiated and proliferative state, whereas the ß-catenin/p300 axis is associated with cell differentiation. We hypothesized that disrupting the ß-catenin/CBP signaling axis would promote epithelial differentiation and inhibit EMT. We treated monolayer cultures of human airway epithelial cells with TGFß1 in the presence or absence of the selective small molecule ICG-001 to inhibit ß-catenin/CBP signaling. We used western blots to assess expression of an EMT signature, CBP, p300, ß-catenin, fibronectin and ITGß1 and scratch wound assays to assess epithelial cell migration. Snai-1 and -2 expressions were determined using q-PCR. Exposure to TGFß1 induced EMT, characterized by reduced E-cadherin expression with increased expression of α-smooth muscle actin and EDA-fibronectin. Either co-treatment or therapeutic administration of ICG-001 completely inhibited TGFß1-induced EMT. ICG-001 also reduced the expression of ck-5 and -19 independent of TGFß1. Exposure to ICG-001 significantly inhibited epithelial cell proliferation and migration, coincident with a down regulation of ITGß1 and fibronectin expression. These data support our hypothesis that modulating the ß-catenin/CBP signaling axis plays a key role in epithelial plasticity and function.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Epithelial Cells/metabolism , Peptide Fragments/genetics , Pyrimidinones/pharmacology , Sialoglycoproteins/genetics , Transforming Growth Factor beta1/pharmacology , beta Catenin/genetics , Actins/genetics , Actins/metabolism , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression Regulation , Humans , Keratin-19/genetics , Keratin-19/metabolism , Keratin-5/genetics , Keratin-5/metabolism , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Primary Cell Culture , Respiratory Mucosa/cytology , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Sialoglycoproteins/antagonists & inhibitors , Sialoglycoproteins/metabolism , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
5.
Am J Respir Cell Mol Biol ; 49(6): 978-88, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23837456

ABSTRACT

The airway epithelium in asthma displays altered repair and incomplete barrier formation. Basal cells are the progenitor cells of the airway epithelium, and can repopulate other cell types after injury. We previously reported increased numbers of basal cells expressing the transcription factor p63 in the airway epithelium of patients with asthma. Here we sought to determine the molecular consequences of p63 expression in basal human airway epithelial cells during wound repair. Because at least six isoforms of p63 exist (N-terminally truncated [ΔN] versus transcriptional activation promoter variants and α, ß, or γ 3' splice variants), the expression of all isoforms was investigated in primary human airway epithelial cells (pHAECs). We modulated p63 expression, using small interfering RNA (siRNA) and adenoviral constructs to determine the effects of p63 on 21 candidate target genes by RT-PCR, and on repair using a scratch wound assay. We found that basal pHAECs from asthmatic and nonasthmatic donors predominantly expressed the N-terminally truncated p63α variant (ΔNp63α) isoform, with no disease-specific differences in expression. The knockdown of ΔNp63, using specific siRNA, decreased the expression of 11 out of 21 genes associated with epithelial repair and differentiation, including ß-catenin, epidermal growth factor receptor, and Jagged1. The loss of ΔNp63 significantly inhibited wound closure (which was associated with the decreased expression of ß-catenin and Jagged1), reduced epithelial proliferation as measured by Ki-67 staining, and increased E-cadherin expression, potentially preventing cytokinesis. In conclusion, ΔNp63α is the major isoform expressed in basal pHAECs, and is essential for epithelial wound repair. The role of ΔNp63α in epithelial barrier integrity requires further study to understand its role in health and disease.


Subject(s)
Respiratory Mucosa/cytology , Respiratory Mucosa/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Wound Healing/genetics , Wound Healing/physiology , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
6.
Hepatology ; 57(6): 2491-501, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23315977

ABSTRACT

MicroRNAs (miRNAs) are recently discovered small RNA molecules that regulate developmental processes, such as proliferation, differentiation, and apoptosis; however, the identity of miRNAs and their functions during liver development are largely unknown. Here we investigated the miRNA and gene expression profiles for embryonic day (E)8.5 endoderm, E14.5 Dlk1(+) liver cells (hepatoblasts), and adult liver by employing Illumina sequencing. We found that miRNAs were abundantly expressed at all three stages. Using K-means clustering analysis, 13 miRNA clusters with distinct temporal expression patterns were identified. mir302b, an endoderm-enriched miRNA, was identified as an miRNA whose predicted targets are expressed highly in E14.5 hepatoblasts but low in the endoderm. We validated the expression of mir302b in the endoderm by whole-mount in situ hybridization. Interestingly, mir20a, the most highly expressed miRNA in the endoderm library, was also predicted to regulate some of the same targets as mir302b. We found that through targeting Tgfbr2, mir302b and mir20a are able to regulate transforming growth factor beta (TGFß) signal transduction. Moreover, mir302b can repress liver markers in an embryonic stem cell differentiation model. Collectively, we uncovered dynamic patterns of individual miRNAs during liver development, as well as miRNA networks that could be essential for the specification and differentiation of liver progenitors. (HEPATOLOGY 2013).


Subject(s)
Liver/embryology , MicroRNAs/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Benzodioxoles/pharmacology , Cell Differentiation , Embryonic Stem Cells/physiology , Endoderm/metabolism , Female , Gastrointestinal Tract/metabolism , Gene Expression Profiling , Genome , Imidazoles/pharmacology , Liver/metabolism , Male , Mice , Organogenesis , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , p300-CBP Transcription Factors/metabolism
7.
J Allergy Clin Immunol ; 129(4): 1116-25.e6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22227418

ABSTRACT

BACKGROUND: The airway epithelium is the first line of defense against inhaled insults and therefore must be capable of coordinating appropriate inflammatory and immune responses. OBJECTIVE: We sought to test the hypothesis that the nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome, an intracellular danger-sensing complex, plays a critical role in airway epithelium-mediated immune responses to urban particulate matter (PM) exposure. METHODS: In this study we (1) identified NLRP3 and caspase-1 expression in human airway epithelium bronchus and primary cells, (2) characterized NLRP3 inflammasome-mediated IL-1ß production from human airway epithelium in response to PM, and (3) performed in vivo PM exposure experiments with wild-type and Nlrp3(-/-) mice. RESULTS: Our results demonstrate that human airway epithelium contains a functional NLRP3 inflammasome that responds to PM exposure with caspase-1 cleavage and production of IL-1ß. Exposure of Nlrp3(-/-) and wild-type mice to PM in vivo demonstrates NLRP3-dependent production of IL-1ß in the lung, airway neutrophilia, and increases in CD11c(+hi)/MHC class II(+hi) cell numbers in intrathoracic lymph nodes. CONCLUSION: Our study is the first to characterize airway epithelial NLRP3 inflammasome-mediated immune responses to PM exposure, which might have implications in patients with asthma and other lung diseases.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Particulate Matter/immunology , Proteins/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Humans , Immunophenotyping , Interleukin-1beta/metabolism , Leucine-Rich Repeat Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Transport , Proteins/genetics
8.
PLoS One ; 3(12): e4039, 2008.
Article in English | MEDLINE | ID: mdl-19112509

ABSTRACT

Bone morphogenic protein (BMP)-7 is a member of the BMP family which are structurally and functionally related, and part of the TGFbeta super family of growth factors. BMP-7 has been reported to inhibit renal fibrosis and TGFbeta1-induced epithelial-mesenchymal transition (EMT), in part through negative interactions with TGFbeta1 induced Smad 2/3 activation. We utilized in vivo bleomycin-induced fibrosis models in the skin and lung to determine the potential therapeutic effect of BMP-7. We then determined the effect of BMP-7 on TGFbeta1-induced EMT in lung epithelial cells and collagen production by human lung fibroblasts. We show that BMP-7 did not affect bleomycin-induced fibrosis in either the lung or skin in vivo; had no effect on expression of pro-fibrotic genes by human lung fibroblasts, either at rest or following exposure to TGFbeta1; and did not modulate TGFbeta1-induced EMT in human lung epithelial cells. Taken together our data indicates that BMP-7 has no anti-fibrotic effect in lung or skin fibrosis either in vivo or in vitro. This suggests that the therapeutic options for BMP-7 may be confined to the renal compartment.


Subject(s)
Bleomycin/pharmacology , Bone Morphogenetic Protein 7/physiology , Fibrosis/chemically induced , Fibrosis/metabolism , Gene Expression Regulation , Lung/pathology , Skin/pathology , Animals , Bone Morphogenetic Protein 7/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Lung/drug effects , Lung/metabolism , Mice , Mice, Inbred C57BL , Skin/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism
9.
Curr Opin Allergy Clin Immunol ; 8(1): 44-8, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18188017

ABSTRACT

PURPOSE OF REVIEW: Asthma remains a severe health problem since current therapies are directed to suppressing, rather than preventing or reversing, the primary disease process. Clearly, a greater understanding of the pathogenesis of asthma is critical to the development of better therapeutic modalities. In this review, we discuss the recent advancements in research targeting the role of airway remodeling in asthma. RECENT FINDINGS: Epithelial fragility and abnormalities are being recognized as important facets of asthma, as are other features of remodeling such as angiogenesis, goblet cell hyperplasia and thickened lamina reticularis. Significantly, these anomalies occur early in disease pathogenesis. However, their impact on disease severity remains unclear. SUMMARY: Although an altered immune response is undoubtedly important to the pathogenesis of asthma, there is increasing evidence that the tissue-specific manifestations occur independently of inflammation and significantly impact on disease development and severity.


Subject(s)
Asthma/pathology , Asthma/physiopathology , Neovascularization, Pathologic , Respiratory System/blood supply , Respiratory System/pathology , ADAM Proteins/genetics , ADAM Proteins/immunology , Animals , Asthma/immunology , Asthma/therapy , Epidermal Growth Factor/immunology , GABA Modulators/therapeutic use , Genetic Therapy , Glucocorticoids/therapeutic use , Goblet Cells/immunology , Goblet Cells/pathology , Humans , Hyperplasia , Mice , Mucous Membrane/pathology , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/pathology , Respiratory System/immunology , alpha Catenin/genetics , alpha Catenin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...