Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Nat Commun ; 14(1): 7349, 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-37963864

ABSTRACT

Toll-like receptor 7 (TLR7) is known for eliciting immunity against single-stranded RNA viruses, and is increased in both human and cigarette smoke (CS)-induced, experimental chronic obstructive pulmonary disease (COPD). Here we show that the severity of CS-induced emphysema and COPD is reduced in TLR7-deficient mice, while inhalation of imiquimod, a TLR7-agonist, induces emphysema without CS exposure. This imiquimod-induced emphysema is reduced in mice deficient in mast cell protease-6, or when wild-type mice are treated with the mast cell stabilizer, cromolyn. Furthermore, therapeutic treatment with anti-TLR7 monoclonal antibody suppresses CS-induced emphysema, experimental COPD and accumulation of pulmonary mast cells in mice. Lastly, TLR7 mRNA is increased in pre-existing datasets from patients with COPD, while TLR7+ mast cells are increased in COPD lungs and associated with severity of COPD. Our results thus support roles for TLR7 in mediating emphysema and COPD through mast cell activity, and may implicate TLR7 as a potential therapeutic target.


Subject(s)
Emphysema , Pulmonary Disease, Chronic Obstructive , Pulmonary Emphysema , Humans , Animals , Mice , Tryptases/genetics , Toll-Like Receptor 7/genetics , Imiquimod , Lung , Pulmonary Emphysema/genetics , Nicotiana , Mice, Inbred C57BL
2.
Am J Clin Pathol ; 160(4): 335-340, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37289435

ABSTRACT

OBJECTIVES: This study aims to determine what pathologic and clinical factors differentiate Brachyspira species that may be useful to clinicians and pathologists. METHODS: We identified 21 studies of Brachyspira infection with individual patient information (n = 113) and conducted a pooled analysis comparing each species. RESULTS: There were differences in the pathologic and clinical profiles of each Brachyspira species. Patients infected with Brachyspira pilosicoli infection were more likely to have diarrhea, fever, HIV, and immunocompromised conditions. Those patients infected with Brachyspira aalborgi were more likely to have lamina propria inflammation. CONCLUSIONS: Our novel data provide potential insights into the pathogenic mechanism(s) and the specific risk factor profile of Brachyspira species. This may be clinically useful when assessing and managing patients.


Subject(s)
Brachyspira , Spirochaetales Infections , Humans , Spirochaetales , Spirochaetales Infections/pathology
3.
J Gastroenterol Hepatol ; 37(7): 1222-1234, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35385602

ABSTRACT

Human colonic spirochetosis (CS) is usually due toBrachyspira pilosicolior Brachyspira aalborgiinfection. While traditionally considered to be commensal bacteria, there are scattered case reports and case series of gastrointestinal (GI) symptoms in CS and reports of colonic polyps with adherent spirochetes. We performed a systematic review and meta-analysis investigating the association between CS and GI symptoms and conditions including the irritable bowel syndrome (IBS) and colonic polyps. Following PRISMA 2020 guidelines, a systematic search of Medline, CINAHL, EMBASE, and Web of Science was performed using specific keywords for CS and GI disease. Pooled odds ratios (ORs) and 95% confidence intervals (CIs) were calculated using a random-effects model. Of 75 studies identified in the search, 8 case-control studies met the inclusion criteria for meta-analysis and 67 case series studies met the inclusion criteria for pooled prevalence analysis. CS was significantly associated with diarrhea (n = 141/127, cases/controls, OR: 4.19, 95% CI: 1.72-10.21, P = 0.002) and abdominal pain (n = 64/65, OR: 3.66, 95% CI: 1.43-9.35, P = 0.007). CS cases were significantly more likely to have Rome III-diagnosed IBS (n = 79/48, OR: 3.84, 95% CI: 1.44-10.20, P = 0.007), but not colonic polyps (n = 127/843, OR: 8.78, 95% CI: 0.75-103.36, P = 0.084). In conclusion, we found evidence of associations between CS and both diarrhea and IBS, but not colonic polyps. CS is likely underestimated due to suboptimal diagnostic methods and may be an overlooked risk factor for a subset of IBS patients with diarrhea.


Subject(s)
Bacterial Infections , Irritable Bowel Syndrome , Diarrhea/etiology , Humans , Intestines , Irritable Bowel Syndrome/diagnosis , Irritable Bowel Syndrome/epidemiology , Prevalence
4.
Brain Behav Immun ; 101: 335-345, 2022 03.
Article in English | MEDLINE | ID: mdl-35093492

ABSTRACT

Functional dyspepsia (FD) affects up to 15% of the population and is characterised by recurring upper gastrointestinal (GI) symptoms occurring in the absence of clinically identifiable pathology. Psychological stress is a key factor associated with the onset of FD and locally acting hypothalamic-pituitary-adrenal (HPA) axis hormones have been implicated in GI motility and barrier dysfunction. Recent pre-clinical work has identified mechanistic pathways linking corticotropin-releasing hormone (CRH) with the innate epithelial immune protein NLRP6, an inflammasome that has been shown to regulate GI mucus secretion. We recruited twelve FD patients and twelve healthy individuals to examine whether dysregulation of hypothalamic-pituitary adrenal (HPA) axis hormones and altered NLRP6 pathways were evident in the duodenal mucosa. Protein expression was assessed by immunoblot and immunohistochemistry in D2 duodenal biopsies. Plasma HPA axis hormones were assayed by ELISA and enteroid and colorectal cancer cell line cultures were used to verify function. FD patients exhibited reduced duodenal CRH-receptor 2, compared to non-GI disease controls, indicating a dysregulation of duodenal HPA signalling. The loss of CRH-receptor 2 correlated with reduced NLRP6 expression and autophagy function, processes critical for maintaining goblet cell homeostasis. In accordance, duodenal goblet cell numbers and mucin exocytosis was reduced in FD patients compared to controls. In vitro studies demonstrated that CRH could reduce NLRP6 in duodenal spheroids and promote mucus secretion in the HT29-MTX-E12 cell line. In conclusion, FD patients exhibit defects in the NLRP6-autophagy axis with decreased goblet cell function that may drive symptoms of disease. These features correlated with loss of CRH receptor 2 and may be driven by dysregulation of HPA signalling in the duodenum of FD patients.


Subject(s)
Dyspepsia , Intracellular Signaling Peptides and Proteins , Pituitary-Adrenal System , Receptors, Corticotropin-Releasing Hormone , Autophagy , Duodenum/metabolism , Dyspepsia/metabolism , Goblet Cells/metabolism , Homeostasis , Hormones/metabolism , Humans , Hypothalamo-Hypophyseal System/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Pituitary-Adrenal System/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
5.
Gastroenterology ; 162(3): 877-889.e7, 2022 03.
Article in English | MEDLINE | ID: mdl-34861219

ABSTRACT

BACKGROUND & AIMS: Excessive shedding of apoptotic enterocytes into the intestinal lumen is observed in inflammatory bowel disease and is correlated with disease relapse. Based on their cytolytic capacity and surveillance behavior, we investigated whether intraepithelial lymphocytes expressing the γδ T cell receptor (γδ IELs) are actively involved in the shedding of enterocytes into the lumen. METHODS: Intravital microscopy was performed on GFP γδ T cell reporter mice treated with intraperitoneal lipopolysaccharide (10 mg/kg) for 90 minutes to induce tumor necrosis factor-mediated apoptosis. Cell shedding in various knockout or transgenic mice in the presence or absence of blocking antibody was quantified by immunostaining for ZO-1 funnels and cleaved caspase-3 (CC3). Granzyme A and granzyme B release from ex vivo-stimulated γδ IELs was quantified by enzyme-linked immunosorbent assay. Immunostaining for γδ T cell receptor and CC3 was performed on duodenal and ileal biopsies from controls and patients with Crohn's disease. RESULTS: Intravital microscopy of lipopolysaccharide-treated mice revealed that γδ IELs make extended contact with shedding enterocytes. These prolonged interactions require CD103 engagement by E-cadherin, and CD103 knockout or blockade significantly reduced lipopolysaccharide-induced shedding. Furthermore, we found that granzymes A and B, but not perforin, are required for cell shedding. These extracellular granzymes are released by γδ IELs both constitutively and after CD103/E-cadherin ligation. Moreover, we found that the frequency of γδ IEL localization to CC3-positive enterocytes is increased in Crohn's disease biopsies compared with healthy controls. CONCLUSIONS: Our results uncover a previously unrecognized role for γδ IELs in facilitating tumor necrosis factor-mediated shedding of apoptotic enterocytes via CD103-mediated extracellular granzyme release.


Subject(s)
Antigens, CD/metabolism , Crohn Disease/metabolism , Enterocytes/physiology , Granzymes/metabolism , Integrin alpha Chains/metabolism , Intraepithelial Lymphocytes/physiology , Adolescent , Adult , Animals , Antigens, CD/genetics , Apoptosis , Cadherins/metabolism , Caspase 3/metabolism , Crohn Disease/pathology , Duodenum/pathology , Enterocytes/metabolism , Female , Gene Knockdown Techniques , Humans , Ileum/pathology , Integrin alpha Chains/genetics , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intraepithelial Lymphocytes/enzymology , Intraepithelial Lymphocytes/pathology , Intravital Microscopy , Jejunum/immunology , Jejunum/pathology , Lipopolysaccharides , Male , Mice , Mice, Knockout , Middle Aged , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Young Adult
6.
Front Immunol ; 13: 1051632, 2022.
Article in English | MEDLINE | ID: mdl-36685573

ABSTRACT

Background: Functional dyspepsia is characterised by chronic symptoms of post-prandial distress or epigastric pain not associated with defined structural pathology. Increased peripheral gut-homing T cells have been previously identified in patients. To date, it is unknown if these T cells were antigen-experienced, or if a specific phenotype was associated with FD. Objective: This study aimed to characterise T cell populations in the blood and duodenal mucosa of FD patients that may be implicated in disease pathophysiology. Methods: We identified duodenal T cell populations from 23 controls and 49 Rome III FD patients by flow cytometry using a surface marker antibody panel. We also analysed T cell populations in peripheral blood from 37 controls and 61 patients. Where available, we examined the number of duodenal eosinophils in patients and controls. Results: There was a shift in the duodenal T helper cell balance in FD patients compared to controls. For example, patients had increased duodenal mucosal Th2 populations in the effector (13.03 ± 16.11, 19.84 ± 15.51, p=0.038), central memory (23.75 ± 18.97, 37.52 ± 17.51, p=0.007) and effector memory (9.80±10.50 vs 20.53±14.15, p=0.001) populations. Th17 populations were also increased in the effector (31.74±24.73 vs 45.57±23.75, p=0.03) and effector memory (11.95±8.42 vs 18.44±15.63, p=0.027) subsets. Peripheral T cell populations were unchanged between FD and control. Conclusion: Our findings identify an association between lymphocyte populations and FD, specifically a Th2 and Th17 signature in the duodenal mucosa. The presence of effector and memory cells suggest that the microinflammation in FD is antigen driven.


Subject(s)
Dyspepsia , Humans , Dyspepsia/diagnosis , Dyspepsia/pathology , Duodenum , Abdominal Pain/metabolism , Eosinophils/metabolism , Mucous Membrane/metabolism
7.
Am J Respir Crit Care Med ; 204(6): 667-681, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34133911

ABSTRACT

Rationale: Necroptosis, mediated by RIPK3 (receptor-interacting protein kinase 3) and MLKL (mixed lineage kinase domain-like), is a form of regulated necrosis that can drive tissue inflammation and destruction; however, its contribution to chronic obstructive pulmonary disease (COPD) pathogenesis is poorly understood. Objectives: To determine the role of necroptosis in COPD. Methods: Total and active (phosphorylated) RIPK3 and MLKL were measured in the lung tissue of patients with COPD and control subjects without COPD. Necroptosis-related mRNA and proteins as well as cell death were examined in lungs and pulmonary macrophages of mice with cigarette smoke (CS)-induced experimental COPD. The responses of Ripk3-/- and Mlkl-/- mice to acute and chronic CS exposure were compared with those of wild-type mice. The combined inhibition of apoptosis (with the pan-caspase inhibitor quinoline-Val-Asp-difluorophenoxymethylketone [qVD-OPh]) and necroptosis (with deletion of Mlkl in mice) was assessed. Measurements and Main Results: The total MLKL protein in the epithelium and macrophages and the pRIPK3 and pMLKL in lung tissue were increased in patients with severe COPD compared with never-smokers or smoker control subjects without COPD. Necroptosis-related mRNA and protein levels were increased in the lungs and macrophages in CS-exposed mice and experimental COPD. Ripk3 or Mlkl deletion prevented airway inflammation upon acute CS exposure. Ripk3 deficiency reduced airway inflammation and remodeling as well as the development of emphysematous pathology after chronic CS exposure. Mlkl deletion and qVD-OPh treatment reduced chronic CS-induced airway inflammation, but only Mlkl deletion prevented airway remodeling and emphysema. Ripk3 or Mlkl deletion and qVD-OPh treatment reduced CS-induced lung-cell death. Conclusions: Necroptosis is induced by CS exposure and is increased in the lungs of patients with COPD and in experimental COPD. Inhibiting necroptosis attenuates CS-induced airway inflammation, airway remodeling, and emphysema. Targeted inhibition of necroptosis is a potential therapeutic strategy in COPD.


Subject(s)
Airway Remodeling , Cigarette Smoking/adverse effects , Inflammation/etiology , Necroptosis , Pulmonary Disease, Chronic Obstructive/physiopathology , Pulmonary Emphysema/etiology , Animals , Case-Control Studies , Disease Progression , Humans , Inflammation/metabolism , Inflammation/physiopathology , Linear Models , Mice , Protein Kinases/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/physiopathology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction
8.
Mucosal Immunol ; 14(2): 366-376, 2021 03.
Article in English | MEDLINE | ID: mdl-32814824

ABSTRACT

In addition to their well characterized role in mediating IgE-dependent allergic diseases, aberrant accumulation and activation of mast cells (MCs) is associated with many non-allergic inflammatory diseases, whereby their activation is likely triggered by non-IgE stimuli (e.g., IL-33). Siglec-8 is an inhibitory receptor expressed on MCs and eosinophils that has been shown to inhibit IgE-mediated MC responses and reduce allergic inflammation upon ligation with a monoclonal antibody (mAb). Herein, we evaluated the effects of an anti-Siglec-8 mAb (anti-S8) in non-allergic disease models of experimental cigarette-smoke-induced chronic obstructive pulmonary disease and bleomycin-induced lung injury in Siglec-8 transgenic mice. Therapeutic treatment with anti-S8 inhibited MC activation and reduced recruitment of immune cells, airway inflammation, and lung fibrosis. Similarly, using a model of MC-dependent, IL-33-induced inflammation, anti-S8 treatment suppressed neutrophil influx, and cytokine production through MC inhibition. Transcriptomic profiling of MCs further demonstrated anti-S8-mediated downregulation of MC signaling pathways induced by IL-33, including TNF signaling via NF-κB. Collectively, these findings demonstrate that ligating Siglec-8 with an antibody reduces non-allergic inflammation and inhibits IgE-independent MC activation, supporting the evaluation of an anti-Siglec-8 mAb as a therapeutic approach in both allergic and non-allergic inflammatory diseases in which MCs play a role.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, B-Lymphocyte/metabolism , Lectins/metabolism , Mast Cells/immunology , Pneumonia/immunology , Pulmonary Disease, Chronic Obstructive/metabolism , Respiratory System/immunology , Animals , Antibodies, Monoclonal/metabolism , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Cell Degranulation , Cigarette Smoking , Disease Models, Animal , Gene Expression Profiling , Immunoglobulin E/metabolism , Interleukin-33/metabolism , Lectins/genetics , Mice , Mice, Transgenic , NF-kappa B/metabolism , Neutrophil Activation , Signal Transduction
9.
FASEB J ; 34(6): 7718-7732, 2020 06.
Article in English | MEDLINE | ID: mdl-32293760

ABSTRACT

Liver inflammation is a common extraintestinal manifestation in inflammatory bowel disease (IBD), yet, the mechanisms driving gut-liver axis inflammation remain poorly understood. IBD leads to a breakdown in the integrity of the intestinal barrier causing an increase in portal and systemic gut-derived antigens, which challenge the liver. Here, we examined the role of platelet activating factor receptor (PAFR) in colitis-associated liver damage using dextran sulfate sodium (DSS) and anti-CD40-induced colitis models. Both DSS and anti-CD40 models exhibited liver inflammation associated with colitis. Colitis reduced global PAFR protein expression in mouse livers causing an exclusive re-localization of PAFR to the portal triad. The global decrease in liver PAFR was associated with increased sirtuin 1 while relocalized PAFR expression was limited to Kupffer cells (KCs) and co-localized with toll-like receptor 4. DSS activated the NLRP3-inflammasome and increased interleukin (IL)-1ß in the liver. Antagonism of PAFR amplified the inflammasome response by increasing NLRP3, caspase-1, and IL-1ß protein levels in the liver. LPS also increased NLRP3 response in human hepatocytes, however, overexpression of PAFR restored the levels of NLPR3 and caspase-1 proteins. Interestingly, KCs depletion also increased IL-1ß protein in mouse liver after DSS challenge. These data suggest a protective role for PAFR-expressing KCs during colitis and that regulation of PAFR is important for gut-liver axis homeostasis.


Subject(s)
Colitis/metabolism , Colitis/pathology , Inflammation/metabolism , Inflammation/pathology , Liver/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Caspase 1/metabolism , Cells, Cultured , Colitis/chemically induced , Colon/metabolism , Colon/pathology , Dextran Sulfate/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Inflammasomes/metabolism , Inflammation/chemically induced , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Interleukin-1beta/metabolism , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Toll-Like Receptor 4/metabolism
10.
JCI Insight ; 52019 07 25.
Article in English | MEDLINE | ID: mdl-31343988

ABSTRACT

Tissue remodeling/fibrosis is a major feature of all fibrotic diseases, including idiopathic pulmonary fibrosis (IPF). It is underpinned by accumulating extracellular matrix (ECM) proteins. Fibulin-1c (Fbln1c) is a matricellular ECM protein associated with lung fibrosis in both humans and mice, and stabilizes collagen formation. Here we discovered that Fbln1c was increased in the lung tissues of IPF patients and experimental bleomycin-induced pulmonary fibrosis. Fbln1c-deficient (-/-) mice had reduced pulmonary remodeling/fibrosis and improved lung function after bleomycin challenge. Fbln1c interacted with fibronectin, periostin and tenascin-c in collagen deposits following bleomycin challenge. In a novel mechanism of fibrosis Fbln1c bound to latent transforming growth factor (TGF)-ß binding protein-1 (LTBP1) to induce TGF-ß activation, and mediated downstream Smad3 phosphorylation/signaling. This process increased myofibroblast numbers and collagen deposition. Fbln1 and LTBP1 co-localized in lung tissues from IPF patients. Thus, Fbln1c may be a novel driver of TGF-ß-induced fibrosis involving LTBP1 and may be an upstream therapeutic target.


Subject(s)
Calcium-Binding Proteins/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Latent TGF-beta Binding Proteins/metabolism , Transforming Growth Factor beta/metabolism , Adult , Animals , Bleomycin/toxicity , Calcium-Binding Proteins/genetics , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/surgery , Lung/cytology , Lung/pathology , Lung Transplantation , Male , Mice , Mice, Knockout , Middle Aged , Primary Cell Culture , Protein Isoforms/metabolism , Young Adult
11.
Clin Transl Immunology ; 8(10): e01084, 2019.
Article in English | MEDLINE | ID: mdl-31921419

ABSTRACT

OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is a progressive disease that causes significant mortality and morbidity worldwide and is primarily caused by the inhalation of cigarette smoke (CS). Lack of effective treatments for COPD means there is an urgent need to identify new therapeutic strategies for the underlying mechanisms of pathogenesis. Tristetraprolin (TTP) encoded by the Zfp36 gene is an anti-inflammatory protein that induces mRNA decay, especially of transcripts encoding inflammatory cytokines, including those implicated in COPD. METHODS: Here, we identify a novel protective role for TTP in CS-induced experimental COPD using Zfp36aa/aa mice, a genetically modified mouse strain in which endogenous TTP cannot be phosphorylated, rendering it constitutively active as an mRNA-destabilising factor. TTP wild-type (Zfp36 +/+) and Zfp36aa/aa active C57BL/6J mice were exposed to CS for four days or eight weeks, and the impact on acute inflammatory responses or chronic features of COPD, respectively, was assessed. RESULTS: After four days of CS exposure, Zfp36aa/aa mice had reduced numbers of airway neutrophils and lymphocytes and mRNA expression levels of cytokines compared to wild-type controls. After eight weeks, Zfp36aa/aa mice had reduced pulmonary inflammation, airway remodelling and emphysema-like alveolar enlargement, and lung function was improved. We then used pharmacological treatments in vivo (protein phosphatase 2A activator, AAL(S), and the proteasome inhibitor, bortezomib) to promote the activation and stabilisation of TTP and show that hallmark features of CS-induced experimental COPD were ameliorated. CONCLUSION: Collectively, our study provides the first evidence for the therapeutic potential of inducing TTP as a treatment for COPD.

12.
Am J Respir Cell Mol Biol ; 58(3): 352-365, 2018 03.
Article in English | MEDLINE | ID: mdl-28960101

ABSTRACT

RelB is a member of the NF-κB family, which is essential for dendritic cell (DC) function and maturation. However, the contribution of RelB to the development of allergic airway inflammation (AAI) is unknown. Here, we identify a pivotal role for RelB in the development of spontaneous AAI that is independent of exogenous allergen exposure. We assessed AAI in two strains of RelB-deficient (RelB-/-) mice: one with a targeted deletion and one expressing a major histocompatibility complex transgene. To determine the importance of RelB in DCs, RelB-sufficient DCs (RelB+/+ or RelB-/-) were adoptively transferred into RelB-/- mice. Both strains had increased pulmonary inflammation compared with their respective wild-type (RelB+/+) and heterozygous (RelB+/-) controls. RelB-/- mice also had increased inflammatory cell influx into the airways, levels of chemokines (CCL2/3/4/5/11/17 and CXCL9/10/13) and T-helper cell type 2-associated cytokines (IL-4/5) in lung tissues, serum IgE, and airway remodeling (mucus-secreting cell numbers, collagen deposition, and epithelial thickening). Transfer of RelB+/- CD11c+ DCs into RelB-/- mice decreased pulmonary inflammation, with reductions in lung chemokines, T-helper cell type 2-associated cytokines (IL-4/5/13/25/33 and thymic stromal lymphopoietin), serum IgE, type 2 innate lymphoid cells, myeloid DCs, γδ T cells, lung Vß13+ T cells, mucus-secreting cells, airway collagen deposition, and epithelial thickening. These data indicate that RelB deficiency may be a key pathway underlying AAI, and that DC-encoded RelB is sufficient to restore control of this inflammation.


Subject(s)
Asthma/immunology , Dendritic Cells/immunology , Pneumonia/immunology , Th2 Cells/immunology , Transcription Factor RelB/genetics , Adoptive Transfer , Airway Remodeling/immunology , Animals , Asthma/pathology , Chemokines/blood , Dendritic Cells/transplantation , Female , Immunoglobulin E/blood , Male , Mice , Mice, Knockout
13.
Am J Physiol Lung Cell Mol Physiol ; 314(2): L298-L317, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29025711

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is the third leading cause of morbidity and death and imposes major socioeconomic burdens globally. It is a progressive and disabling condition that severely impairs breathing and lung function. There is a lack of effective treatments for COPD, which is a direct consequence of the poor understanding of the underlying mechanisms involved in driving the pathogenesis of the disease. Toll-like receptor (TLR)2 and TLR4 are implicated in chronic respiratory diseases, including COPD, asthma and pulmonary fibrosis. However, their roles in the pathogenesis of COPD are controversial and conflicting evidence exists. In the current study, we investigated the role of TLR2 and TLR4 using a model of cigarette smoke (CS)-induced experimental COPD that recapitulates the hallmark features of human disease. TLR2, TLR4, and associated coreceptor mRNA expression was increased in the airways in both experimental and human COPD. Compared with wild-type (WT) mice, CS-induced pulmonary inflammation was unaltered in TLR2-deficient ( Tlr2-/-) and TLR4-deficient ( Tlr4-/-) mice. CS-induced airway fibrosis, characterized by increased collagen deposition around small airways, was not altered in Tlr2-/- mice but was attenuated in Tlr4-/- mice compared with CS-exposed WT controls. However, Tlr2-/- mice had increased CS-induced emphysema-like alveolar enlargement, apoptosis, and impaired lung function, while these features were reduced in Tlr4-/- mice compared with CS-exposed WT controls. Taken together, these data highlight the complex roles of TLRs in the pathogenesis of COPD and suggest that activation of TLR2 and/or inhibition of TLR4 may be novel therapeutic strategies for the treatment of COPD.


Subject(s)
Emphysema/etiology , Nicotiana/toxicity , Pneumonia/etiology , Pulmonary Disease, Chronic Obstructive/pathology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Animals , Apoptosis , Bronchoalveolar Lavage Fluid , Emphysema/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/metabolism
14.
J Pathol ; 243(4): 510-523, 2017 12.
Article in English | MEDLINE | ID: mdl-28862768

ABSTRACT

Asthma is a chronic inflammatory disease of the airways. It is characterized by allergic airway inflammation, airway remodelling, and airway hyperresponsiveness (AHR). Asthma patients, in particular those with chronic or severe asthma, have airway remodelling that is associated with the accumulation of extracellular matrix (ECM) proteins, such as collagens. Fibulin-1 (Fbln1) is an important ECM protein that stabilizes collagen and other ECM proteins. The level of Fbln1c, one of the four Fbln1 variants, which predominates in both humans and mice, is increased in the serum and airways fluids in asthma but its function is unclear. We show that the level of Fbln1c was increased in the lungs of mice with house dust mite (HDM)-induced chronic allergic airway disease (AAD). Genetic deletion of Fbln1c and therapeutic inhibition of Fbln1c in mice with chronic AAD reduced airway collagen deposition, and protected against AHR. Fbln1c-deficient (Fbln1c-/- ) mice had reduced mucin (MUC) 5 AC levels, but not MUC5B levels, in the airways as compared with wild-type (WT) mice. Fbln1c interacted with fibronectin and periostin that was linked to collagen deposition around the small airways. Fbln1c-/- mice with AAD also had reduced numbers of α-smooth muscle actin-positive cells around the airways and reduced airway contractility as compared with WT mice. After HDM challenge, these mice also had fewer airway inflammatory cells, reduced interleukin (IL)-5, IL-13, IL-33, tumour necrosis factor (TNF) and CXCL1 levels in the lungs, and reduced IL-5, IL-33 and TNF levels in lung-draining lymph nodes. Therapeutic targeting of Fbln1c reduced the numbers of GATA3-positive Th2 cells in the lymph nodes and lungs after chronic HDM challenge. Treatment also reduced the secretion of IL-5 and IL-13 from co-cultured dendritic cells and T cells restimulated with HDM extract. Human epithelial cells cultured with Fbln1c peptide produced more CXCL1 mRNA than medium-treated controls. Our data show that Fbln1c may be a therapeutic target in chronic asthma. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Airway Remodeling , Asthma/metabolism , Bronchial Hyperreactivity/metabolism , Calcium-Binding Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Inflammation/metabolism , Lung/metabolism , Actins/metabolism , Animals , Asthma/immunology , Asthma/physiopathology , Asthma/prevention & control , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchial Hyperreactivity/prevention & control , Bronchoconstriction , Calcium-Binding Proteins/deficiency , Calcium-Binding Proteins/genetics , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/genetics , Female , Genotype , Humans , Inflammation/immunology , Inflammation/physiopathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Lung/immunology , Lung/physiopathology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA Interference , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transfection
15.
JCI Insight ; 2(7): e90443, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28405612

ABSTRACT

Influenza A virus (IAV) infections lead to severe inflammation in the airways. Patients with chronic obstructive pulmonary disease (COPD) characteristically have exaggerated airway inflammation and are more susceptible to infections with severe symptoms and increased mortality. The mechanisms that control inflammation during IAV infection and the mechanisms of immune dysregulation in COPD are unclear. We found that IAV infections lead to increased inflammatory and antiviral responses in primary bronchial epithelial cells (pBECs) from healthy nonsmoking and smoking subjects. In pBECs from COPD patients, infections resulted in exaggerated inflammatory but deficient antiviral responses. A20 is an important negative regulator of NF-κB-mediated inflammatory but not antiviral responses, and A20 expression was reduced in COPD. IAV infection increased the expression of miR-125a or -b, which directly reduced the expression of A20 and mitochondrial antiviral signaling (MAVS), and caused exaggerated inflammation and impaired antiviral responses. These events were replicated in vivo in a mouse model of experimental COPD. Thus, miR-125a or -b and A20 may be targeted therapeutically to inhibit excessive inflammatory responses and enhance antiviral immunity in IAV infections and in COPD.


Subject(s)
Influenza, Human/immunology , MicroRNAs/genetics , Mitochondria/immunology , Proteins/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Aged , Animals , Case-Control Studies , Cells, Cultured , Epithelial Cells/immunology , Female , Humans , Inflammation/etiology , Influenza A virus/physiology , Influenza, Human/complications , Intracellular Signaling Peptides and Proteins , Male , Mice, Inbred BALB C , Middle Aged , NF-kappa B/metabolism , Pulmonary Disease, Chronic Obstructive/complications , Smoking/adverse effects , Virus Replication
16.
J Allergy Clin Immunol ; 139(2): 519-532, 2017 02.
Article in English | MEDLINE | ID: mdl-27448447

ABSTRACT

BACKGROUND: Severe steroid-insensitive asthma is a substantial clinical problem. Effective treatments are urgently required, however, their development is hampered by a lack of understanding of the mechanisms of disease pathogenesis. Steroid-insensitive asthma is associated with respiratory tract infections and noneosinophilic endotypes, including neutrophilic forms of disease. However, steroid-insensitive patients with eosinophil-enriched inflammation have also been described. The mechanisms that underpin infection-induced, severe steroid-insensitive asthma can be elucidated by using mouse models of disease. OBJECTIVE: We sought to develop representative mouse models of severe, steroid-insensitive asthma and to use them to identify pathogenic mechanisms and investigate new treatment approaches. METHODS: Novel mouse models of Chlamydia, Haemophilus influenzae, influenza, and respiratory syncytial virus respiratory tract infections and ovalbumin-induced, severe, steroid-insensitive allergic airway disease (SSIAAD) in BALB/c mice were developed and interrogated. RESULTS: Infection induced increases in the levels of microRNA (miRNA)-21 (miR-21) expression in the lung during SSIAAD, whereas expression of the miR-21 target phosphatase and tensin homolog was reduced. This was associated with an increase in levels of phosphorylated Akt, an indicator of phosphoinositide 3-kinase (PI3K) activity, and decreased nuclear histone deacetylase (HDAC)2 levels. Treatment with an miR-21-specific antagomir (Ant-21) increased phosphatase and tensin homolog levels. Treatment with Ant-21, or the pan-PI3K inhibitor LY294002, reduced PI3K activity and restored HDAC2 levels. This led to suppression of airway hyperresponsiveness and restored steroid sensitivity to allergic airway disease. These observations were replicated with SSIAAD associated with 4 different pathogens. CONCLUSION: We identify a previously unrecognized role for an miR-21/PI3K/HDAC2 axis in SSIAAD. Our data highlight miR-21 as a novel therapeutic target for the treatment of this form of asthma.


Subject(s)
Asthma/genetics , Chlamydia muridarum/immunology , Haemophilus influenzae/immunology , Histone Deacetylase 2/metabolism , Influenza A Virus, H1N1 Subtype/immunology , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Pneumonia/genetics , Respiratory Syncytial Viruses/immunology , Respiratory Tract Infections/genetics , Animals , Antagomirs/genetics , Asthma/drug therapy , Asthma/immunology , Dexamethasone/therapeutic use , Disease Models, Animal , Drug Resistance , Gene Expression Regulation , Histone Deacetylase 2/genetics , Humans , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Pneumonia/drug therapy , Pneumonia/immunology , Proto-Oncogene Proteins c-akt/metabolism , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology
17.
JCI Insight ; 1(9)2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27398409

ABSTRACT

Airway and/or lung remodeling, involving exaggerated extracellular matrix (ECM) protein deposition, is a critical feature common to pulmonary diseases including chronic obstructive pulmonary disease (COPD), asthma, and idiopathic pulmonary fibrosis (IPF). Fibulin-1 (Fbln1), an important ECM protein involved in matrix organization, may be involved in the pathogenesis of these diseases. We found that Fbln1 was increased in COPD patients and in cigarette smoke-induced (CS-induced) experimental COPD in mice. Genetic or therapeutic inhibition of Fbln1c protected against CS-induced airway fibrosis and emphysema-like alveolar enlargement. In experimental COPD, this occurred through disrupted collagen organization and interactions with fibronectin, periostin, and tenascin-c. Genetic inhibition of Fbln1c also reduced levels of pulmonary inflammatory cells and proinflammatory cytokines/chemokines (TNF-α, IL-33, and CXCL1) in experimental COPD. Fbln1c-/- mice also had reduced airway remodeling in experimental chronic asthma and pulmonary fibrosis. Our data show that Fbln1c may be a therapeutic target in chronic respiratory diseases.

18.
J Immunol ; 191(8): 4112-20, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24048894

ABSTRACT

The induction of regulatory T cells (Tregs) to suppress aberrant inflammation and immunity has potential as a therapeutic strategy for asthma. Recently, we identified key immunoregulatory components of Streptococcus pneumoniae, type 3 polysaccharide and pneumolysoid (T+P), which suppress allergic airways disease (AAD) in mouse models of asthma. To elucidate the mechanisms of suppression, we have now performed a thorough examination of the role of Tregs. BALB/c mice were sensitized to OVA (day 0) i.p. and challenged intranasal (12-15 d later) to induce AAD. T+P was administered intratracheally at the time of sensitization in three doses (0, 12, and 24 h). T+P treatment induced an early (36 h-4 d) expansion of Tregs in the mediastinal lymph nodes, and later (12-16 d) increases in these cells in the lungs, compared with untreated allergic controls. Anti-CD25 treatment showed that Treg-priming events involving CD25, CCR7, IL-2, and TGF-ß were required for the suppression of AAD. During AAD, T+P-induced Tregs in the lungs displayed a highly suppressive phenotype and had an increased functional capacity. T+P also blocked the induction of IL-6 to prevent the Th17 response, attenuated the expression of the costimulatory molecule CD86 on myeloid dendritic cells (DCs), and reduced the number of DCs carrying OVA in the lung and mediastinal lymph nodes. Therefore, bacterial components (T+P) drive the differentiation of highly suppressive Tregs, which suppress the Th2 response, prevent the Th17 response and disable the DC response resulting in the effective suppression of AAD.


Subject(s)
Asthma/immunology , Polysaccharides, Bacterial/immunology , Streptococcus pneumoniae/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-2 Antigen/biosynthesis , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Inflammation/immunology , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-6/biosynthesis , Lung/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Polysaccharides, Bacterial/administration & dosage , Receptors, CCR7/metabolism , Th17 Cells/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/metabolism
19.
Clin Obstet Gynecol ; 56(3): 566-76, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23722921

ABSTRACT

There is emerging evidence that chronic respiratory diseases such as asthma and emphysema may originate in early life. Respiratory infections with certain bacterial pathogens such as Chlamydia, Haemophilus influenzae and Streptococcus pneumoniae in early life may promote permanent deleterious changes in immunity, lung structure, and function that predispose to, or increase the severity of chronic respiratory diseases in later life. For example, these infections increase immune responses, which drive subsequent asthma pathogenesis. Targeting the pathways involved with specific inhibitors or agonists may prevent these consequences of early-life infection. Vaccination and immunomodulatory therapies that control the infections and their sequelae may also be efficacious.


Subject(s)
Asthma/immunology , Bacterial Infections/immunology , Lung/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Respiratory Tract Infections/immunology , Asthma/etiology , Bacterial Infections/drug therapy , Bacterial Infections/prevention & control , Chlamydia Infections/drug therapy , Chlamydia Infections/immunology , Chlamydia trachomatis , Chlamydophila pneumoniae , Haemophilus Infections/drug therapy , Haemophilus Infections/immunology , Haemophilus influenzae , Humans , Immunologic Factors/therapeutic use , Interleukin-13/antagonists & inhibitors , Pulmonary Disease, Chronic Obstructive/etiology , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/prevention & control , Streptococcal Infections/drug therapy , Streptococcal Infections/immunology , Streptococcal Infections/prevention & control , Streptococcal Vaccines/therapeutic use , Streptococcus pneumoniae , Toll-Like Receptor 2/agonists
SELECTION OF CITATIONS
SEARCH DETAIL
...