Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Sci Rep ; 11(1): 15918, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354132

ABSTRACT

Acetylcholine (ACh), the neurotransmitter of the cholinergic system, regulates inflammation in several diseases including pulmonary diseases. ACh is also involved in a non-neuronal mechanism that modulates the innate immune response. Because inflammation and release of pro-inflammatory cytokines are involved in pulmonary emphysema, we hypothesized that vesicular acetylcholine transport protein (VAChT) deficiency, which leads to reduction in ACh release, can modulate lung inflammation in an experimental model of emphysema. Mice with genetical reduced expression of VAChT (VAChT KDHOM 70%) and wild-type mice (WT) received nasal instillation of 50 uL of porcine pancreatic elastase (PPE) or saline on day 0. Twenty-eight days after, animals were evaluated. Elastase instilled VAChT KDHOM mice presented an increase in macrophages, lymphocytes, and neutrophils in bronchoalveolar lavage fluid and MAC2-positive macrophages in lung tissue and peribronchovascular area that was comparable to that observed in WT mice. Conversely, elastase instilled VAChT KDHOM mice showed significantly larger number of NF-κB-positive cells and isoprostane staining in the peribronchovascular area when compared to elastase-instilled WT-mice. Moreover, elastase-instilled VAChT-deficient mice showed increased MCP-1 levels in the lungs. Other cytokines, extracellular matrix remodeling, alveolar enlargement, and lung function were not worse in elastase-instilled VAChT deficiency than in elastase-instilled WT-controls. These data suggest that decreased VAChT expression may contribute to the pathogenesis of emphysema, at least in part, through NF-κB activation, MCP-1, and oxidative stress pathways. This study highlights novel pathways involved in lung inflammation that may contribute to the development of chronic obstrutive lung disease (COPD) in cholinergic deficient individuals such as Alzheimer's disease patients.


Subject(s)
Acetylcholine/deficiency , Emphysema/immunology , Pneumonia/etiology , Acetylcholine/metabolism , Animals , Bronchoalveolar Lavage Fluid/cytology , Cytokines/metabolism , Disease Models, Animal , Emphysema/metabolism , Inflammation/pathology , Lung/pathology , Macrophages/metabolism , Male , Mice , NF-kappa B/metabolism , Neutrophils/metabolism , Pancreatic Elastase/adverse effects , Pancreatic Elastase/pharmacology , Pneumonia/physiopathology , Pulmonary Emphysema/metabolism , Signal Transduction , Vesicular Acetylcholine Transport Proteins/deficiency , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism
2.
Inflamm Res ; 69(4): 423-434, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32144443

ABSTRACT

OBJECTIVE AND DESIGN: Gallic acid (GA) a naturally occurring phenolic compound, known to possess antioxidant/anti-inflammatory activities. The aim of the present work was to investigate the beneficial effects of GA against COPD-linked lung inflammation/emphysema by utilizing elastase (ET) and cigarette smoke (CS)-induced mice model. MATERIALS: Male BALB/c mice were treated with ET (1U/mouse) or exposed to CS (9 cigarettes/day for 4 days). GA administration was started 7 days (daily) prior to ET/CS exposure. Broncho-alveolar lavage was analyzed for inflammatory cells and pro-inflammatory cytokines. Lung homogenate was assessed for MPO activity/GSH/MDA/protein carbonyls. Further, Lung tissue was subjected to semi-quantitative RT-PCR, immunoblotting, and histological analysis. RESULTS: GA suppressed the ET-induced neutrophil infiltration, elevated MPO activity and production of pro-inflammatory cytokines (IL-6/TNF-α/IL-1ß) at 24 h. Reduced inflammation was accompanied with normalization of redox balance as reflected by ROS/GSH/MDA/protein carbonyl levels. Further, GA suppressed phosphorylation of p65NF-κB and IκBα along with down-regulation of IL-1ß/TNF-α/KC/MIP-2/GCSF genes. Furthermore, GA offered protection against ET-induced airspace enlargement and ameliorated MMP-2/MMP-9. Finally, GA suppressed the CS-induced influx of neutrophils and macrophages and blunted gene expression of TNF-α/MIP-2/KC. CONCLUSION: Overall, our data show that GA effectively modulates pulmonary inflammation and emphysema associated with COPD pathogenesis in mice.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Emphysema/drug therapy , Gallic Acid/therapeutic use , Pulmonary Disease, Chronic Obstructive/drug therapy , Animals , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/immunology , Emphysema/genetics , Emphysema/immunology , Lung/drug effects , Lung/immunology , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mice, Inbred BALB C , NF-kappa B/immunology , Pancreatic Elastase , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/immunology , Smoke/adverse effects , Tissue Inhibitor of Metalloproteinase-1/genetics , Nicotiana
3.
Front Immunol ; 10: 1323, 2019.
Article in English | MEDLINE | ID: mdl-31244859

ABSTRACT

Microfold (M) cells residing in the follicle-associated epithelium of mucosa-associated lymphoid tissues are specialized for sampling luminal antigens to initiate mucosal immune responses. In the past decade, glycoprotein 2 (GP2) and Tnfaip2 were identified as reliable markers for M cells in the Peyer's patches of the intestine. Furthermore, RANKL-RANK signaling, as well as the canonical and non-canonical NFκB pathways downstream, is essential for M-cell differentiation from the intestinal stem cells. However, the molecular characterization and differentiation mechanisms of M cells in the lower respiratory tract, where organized lymphoid tissues exist rarely, remain to be fully elucidated. Therefore, this study aimed to explore M cells in the lower respiratory tract in terms of their specific molecular markers, differentiation mechanism, and functions. Immunofluorescence analysis revealed a small number of M cells expressing GP2, Tnfaip2, and RANK is present in the lower respiratory tract of healthy mice. The intraperitoneal administration of RANKL in mice effectively induced M cells, which have a high capacity to take up luminal substrates, in the lower respiratory epithelium. The airway M cells associated with lymphoid follicles were frequently detected in the pathologically induced bronchus-associated lymphoid tissue (iBALT) in the murine models of autoimmune disease as well as pulmonary emphysema. These findings demonstrate that RANKL is a common inducer of M cells in the airway and digestive tracts and that M cells are associated with the respiratory disease. We also established a two-dimensional culture method for airway M cells from the tracheal epithelium in the presence of RANKL successfully. This model may be useful for functional studies of M cells in the sampling of antigens at airway mucosal surfaces.


Subject(s)
Immunity, Mucosal , RANK Ligand/immunology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Tract Diseases/immunology , Respiratory Tract Diseases/pathology , Animals , Bronchioles/immunology , Bronchioles/pathology , Cell Culture Techniques , Cigarette Smoking/adverse effects , Cigarette Smoking/immunology , Cigarette Smoking/pathology , Disease Models, Animal , Emphysema/immunology , Emphysema/pathology , Female , GPI-Linked Proteins/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Receptor Activator of Nuclear Factor-kappa B/immunology , Signal Transduction/immunology , Tumor Necrosis Factors/immunology
4.
JCI Insight ; 52019 05 21.
Article in English | MEDLINE | ID: mdl-31112138

ABSTRACT

Alteration of innate immune cells in the lungs can promote loss of peripheral tolerance that leads to autoimmune responses in cigarette smokers. Development of autoimmunity in smokers with emphysema is also strongly linked to the expansion of autoreactive T helper (Th) cells expressing interferon gamma (Th1), and interleukin 17A (Th17). However, the mechanisms responsible for enhanced self-recognition and reduced immune tolerance in smoker with emphysema remain less clear. Here we show that C1q, a component of the complement protein 1 complex (C1), is downregulated in lung CD1a+ antigen presenting cells (APCs) isolated from emphysematous human, and mouse lung APCs after chronic cigarette smoke exposure. C1q potentiated the function of APCs to differentiate CD4+ T cells to Tregs, while it inhibited Th17 cell development and proliferation. Mice deficient in C1q that were exposed to chronic smoke exhibited exaggerated lung inflammation marked by increased Th17 cells, while reconstitution of C1q in the lungs enhanced Tregs abundance, dampened smoke-induced lung inflammation, and reversed established emphysema. Our findings demonstrate that cigarette smoke-mediated loss of C1q could play a key role in reduced peripheral tolerance, which could be explored to treat emphysema.


Subject(s)
Antigen-Presenting Cells/metabolism , Cigarette Smoking/adverse effects , Complement C1q/metabolism , Emphysema/immunology , Th17 Cells/immunology , Adult , Aged , Animals , Antigen-Presenting Cells/immunology , Autoimmunity , Case-Control Studies , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Cigarette Smoking/immunology , Coculture Techniques , Complement C1q/genetics , Complement C1q/immunology , Disease Models, Animal , Down-Regulation/immunology , Emphysema/pathology , Female , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Immune Tolerance , Lung/cytology , Lung/immunology , Lung/pathology , Lymphocyte Activation , Male , Mice , Mice, Knockout , Middle Aged , Primary Cell Culture , Smoke/adverse effects , T-Lymphocytes, Regulatory/immunology , Tissue Array Analysis , Tobacco Products/adverse effects
5.
Transpl Infect Dis ; 21(1): e13026, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30414224

ABSTRACT

Emphysematous pyelonephritis (EPN) is a rare condition which can rapidly progress to sepsis and multiple organ failure with high mortality. We experienced a rare case of EPN in a renal allograft related to antibody-mediated rejection (AMR). The patient received a deceased donor kidney transplant due to end-stage renal disease secondary to diabetes mellitus. Cross-match test was negative but she had remote history of anti-HLA-A2 antibody corresponding with the donor HLA. Surgery concluded without any major events. Anti-thymoglobulin was given perioperatively for induction. She was compliant with her immunosuppressive medications making urine of 2 L/d with serum creatinine of 1.9 mg/dL at discharge on post-operative day (POD) 6. She did well until POD 14 when she presented to the clinic with features of sepsis, pain over the transplanted kidney area and decline in urine volume with elevated serum creatinine. CT revealed extensive gas throughout the transplanted kidney. Renal scan revealed non-functional transplant kidney with no arterial flow. Based on these findings, a decision to perform transplant nephrectomy was made. At laparotomy, the kidney was completely necrotic. Pathology showed non-viable kidney parenchyma with the tubules lacking neutrophilic casts suggestive of ischemic necrosis. Donor-specific antibody (DSA) returned positive with high intensity anti-HLA-A2 antibody. This is the first case of early EPN in allograft considered to have occurred as a result of thrombotic ischemia secondary to AMR. This case suggests consideration of perioperative anti-B-cell and/or anti-plasma cell therapies for historical DSA and strict post-operative follow-up in immunologically high-risk recipients to detect early signs of rejection and avoid deleterious outcomes.


Subject(s)
Emphysema/immunology , Graft Rejection/immunology , Isoantibodies/immunology , Kidney Transplantation/adverse effects , Pyelonephritis/immunology , Allografts/blood supply , Allografts/diagnostic imaging , Allografts/immunology , Allografts/pathology , Biopsy , Emphysema/diagnosis , Emphysema/pathology , Emphysema/therapy , Female , Graft Rejection/diagnosis , Graft Rejection/pathology , Graft Rejection/therapy , Graft Survival/immunology , Humans , Ischemia/diagnosis , Ischemia/immunology , Ischemia/pathology , Ischemia/therapy , Kidney/blood supply , Kidney/diagnostic imaging , Kidney/immunology , Kidney/pathology , Kidney Failure, Chronic/surgery , Middle Aged , Pyelonephritis/diagnosis , Pyelonephritis/pathology , Pyelonephritis/therapy , Radioisotope Renography , Renal Dialysis , Thromboembolism/diagnosis , Thromboembolism/immunology , Thromboembolism/pathology , Thromboembolism/therapy
6.
Cell Rep ; 25(10): 2775-2783.e3, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30517865

ABSTRACT

Emphysema results in destruction of alveolar walls and enlargement of lung airspaces and has been shown to develop during helminth infections through IL-4R-independent mechanisms. We examined whether interleukin 17A (IL-17A) may instead modulate development of emphysematous pathology in mice infected with the helminth parasite Nippostrongylus brasiliensis. We found that transient elevations in IL-17A shortly after helminth infection triggered subsequent emphysema that destroyed alveolar structures. Furthermore, lung B cells, activated through IL-4R signaling, inhibited early onset of emphysematous pathology. IL-10 and other regulatory cytokines typically associated with B regulatory cell function did not play a major role in this response. Instead, at early stages of the response, B cells produced high levels of the tissue-protective protein, Resistin-like molecule α (RELMα), which then downregulated IL-17A expression. These studies show that transient elevations in IL-17A trigger emphysema and reveal a helminth-induced immune regulatory mechanism that controls IL-17A and the severity of emphysema.


Subject(s)
B-Lymphocytes/metabolism , Emphysema/immunology , Emphysema/parasitology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-17/metabolism , Nippostrongylus/physiology , Strongylida Infections/parasitology , Acute Lung Injury/complications , Acute Lung Injury/immunology , Animals , Antibodies/pharmacology , Down-Regulation , Immunity/drug effects , Lung/immunology , Lung/parasitology , Lung/pathology , Mice, Inbred BALB C , Phenotype , Receptors, Interleukin-4/metabolism , Signal Transduction
7.
J Agric Food Chem ; 66(29): 7643-7654, 2018 Jul 25.
Article in English | MEDLINE | ID: mdl-29945446

ABSTRACT

Airway inflammation has been implicated in evoking progressive pulmonary disorders including chronic obstructive pulmonary disease (COPD) and asthma as a result of exposure to inhaled irritants, characterized by airway fibrosis, mucus hypersecretion, and loss of alveolar integrity. The current study examined whether oleuropein, a phenylethanoid found in olive leaves, inhibited pulmonary inflammation in experimental models of interleukin (IL)-4-exposed bronchial BEAS-2B epithelial cells and ovalbumin (OVA)- or cigarette smoke (CS)-exposed BALB/c mice. Nontoxic oleuropein at 1-20 µM diminished eotaxin-1-mediated induction of α-smooth muscle actin and mucin 5AC in epithelial cells stimulated by IL-4 at the transcriptional levels. Oral supplementation of 10-20 mg/kg oleuropein reduced the airway influx of eosinophils and lymphocytes as well as IL-4 secretion in lung promoted by OVA inhalation or CS. In addition, oleuropein suppressed infiltration of macrophages and neutrophils through blocking OVA inhalation- and CS-promoted induction of ICAM-1, F4/80, CD68, and CD11b in airways. OVA-exposed pulmonary fibrosis was detected, while alveolar emphysema was evident in CS-exposed mouse lungs. In alveolar epithelial A549 cells exposed to CS extracts, oleuropein attenuated apoptotic cell loss. Collectively, oleuropein inhibited pulmonary inflammation leading to asthmatic fibrosis and alveolar emphysema driven by influx of inflammatory cells in airways exposed OVA or CS. Therefore, oleuropein may be a promising anti-inflammatory agent for treating asthma and COPD.


Subject(s)
Asthma/drug therapy , Emphysema/drug therapy , Iridoids/administration & dosage , Pneumonia/drug therapy , Animals , Apoptosis/drug effects , Asthma/etiology , Asthma/genetics , Asthma/immunology , Cigarette Smoking/adverse effects , Emphysema/etiology , Emphysema/genetics , Emphysema/immunology , Humans , Immunoglobulin E/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Iridoid Glucosides , Male , Mice , Mice, Inbred BALB C , Pneumonia/etiology , Pneumonia/genetics , Pneumonia/immunology
8.
J Immunol Res ; 2017: 7915975, 2017.
Article in English | MEDLINE | ID: mdl-28536707

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a major cause of mortality worldwide, which is characterized by chronic bronchitis, destruction of small airways, and enlargement/disorganization of alveoli. It is generally accepted that the neutrophilic airway inflammation observed in the lungs of COPD patients is intrinsically linked to the tissue destruction and alveolar airspace enlargement, leading to disease progression. Animal models play an important role in studying the underlying mechanisms of COPD as they address questions involving integrated whole body responses. This review aims to summarize the current animal models of COPD, focusing on their advantages and disadvantages on immune responses and neutrophilic inflammation. Also, we propose a potential new animal model of COPD, which may mimic the most characteristics of human COPD pathogenesis, including persistent moderate-to-high levels of neutrophilic inflammation.


Subject(s)
Disease Models, Animal , Inflammation , Neutrophils/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Animals , Disease Progression , Emphysema/immunology , Emphysema/physiopathology , Humans , Lung/pathology , Mice , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/physiopathology , Rats , Smoking
9.
J Immunol ; 198(9): 3637-3649, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28363903

ABSTRACT

Acute exacerbation of chronic obstructive pulmonary disease (COPD) is often induced by infection and often has a poor prognosis. Bacterial LPS activates innate immune receptor TLR4 followed by activation of a transcriptional factor IFN regulatory factor-3 (IRF3) as well as NF-κB, resulting in upregulation of various inflammatory mediators. To clarify the role of IRF3 in the pathogenesis of LPS-triggered COPD exacerbation, porcine pancreatic elastase (PPE) followed by LPS was administered intranasally to wild-type (WT) or IRF3-/- male mice. Sequential quantitative changes in emphysema were evaluated by microcomputed tomography, and lung histology was evaluated at the sixth week. WT mice treated with PPE and LPS exhibited enlarged alveolar spaces, whereas this feature was attenuated in similarly treated IRF3-/- mice. Moreover, LPS-induced emphysema aggravation was detected only in WT mice. Analysis of acute inflammation induced by PPE plus LPS revealed that the lungs of treated IRF3-/- mice had decreased mRNA transcripts for MCP-1, MIP-1α, TNF-α, and IFN-γ-inducible protein-10 but had increased neutrophils. IRF3 was involved in the production of mediators from macrophages, alveolar epithelial cells, and neutrophils. Furthermore, compared with isolated WT neutrophils from inflamed lung, those of IRF3-/- neutrophils exhibited impaired autophagic activation, phagocytosis, and apoptosis. These results suggest that IRF3 accelerated emphysema formation based on distinct profiles of mediators involved in LPS-induced COPD exacerbation. Regulation of the IRF3 pathway can affect multiple cell types and contribute to ameliorate pathogenesis of infection-triggered exacerbation of COPD.


Subject(s)
Emphysema/immunology , Interferon Regulatory Factor-3/metabolism , Lung/immunology , Neutrophils/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Animals , Apoptosis/genetics , Autophagy/genetics , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Interferon-gamma/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
10.
PLoS One ; 12(3): e0173446, 2017.
Article in English | MEDLINE | ID: mdl-28291826

ABSTRACT

Endothelial injury is implicated in the pathogenesis of COPD and emphysema; however the role of endothelial progenitor cells (EPCs), a marker of endothelial cell repair, and circulating endothelial cells (CECs), a marker of endothelial cell injury, in COPD and its subphenotypes is unresolved. We hypothesized that endothelial progenitor cell populations would be decreased in COPD and emphysema and that circulating endothelial cells would be increased. Associations with other subphenotypes were examined. The Multi-Ethnic Study of Atherosclerosis COPD Study recruited smokers with COPD and controls age 50-79 years without clinical cardiovascular disease. Endothelial progenitor cell populations (CD34+KDR+ and CD34+KDR+CD133+ cells) and circulating endothelial cells (CD45dimCD31+CD146+CD133-) were measured by flow cytometry. COPD was defined by standard spirometric criteria. Emphysema was assessed qualitatively and quantitatively on CT. Full pulmonary function testing and expiratory CTs were measured in a subset. Among 257 participants, both endothelial progenitor cell populations, and particularly CD34+KDR+ endothelial progenitor cells, were reduced in COPD. The CD34+KDR+CD133+ endothelial progenitor cells were associated inversely with emphysema extent. Both endothelial progenitor cell populations were associated inversely with extent of panlobular emphysema and positively with diffusing capacity. Circulating endothelial cells were not significantly altered in COPD but were inversely associated with pulmonary microvascular blood flow on MRI. There was no consistent association of endothelial progenitor cells or circulating endothelial cells with measures of gas trapping. These data provide evidence that endothelial repair is impaired in COPD and suggest that this pathological process is specific to emphysema.


Subject(s)
Emphysema/pathology , Endothelial Progenitor Cells/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Aged , Emphysema/immunology , Endothelial Progenitor Cells/immunology , Female , Flow Cytometry , Humans , Immunophenotyping , Magnetic Resonance Imaging , Male , Middle Aged , Plethysmography , Pulmonary Disease, Chronic Obstructive/immunology
11.
Nat Immunol ; 16(11): 1185-94, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26437241

ABSTRACT

Smoking-related emphysema is a chronic inflammatory disease driven by the T(H)17 subset of helper T cells through molecular mechanisms that remain obscure. Here we explored the role of the microRNA miR-22 in emphysema. We found that miR-22 was upregulated in lung myeloid dendritic cells (mDCs) of smokers with emphysema and antigen-presenting cells (APCs) of mice exposed to smoke or nanoparticulate carbon black (nCB) through a mechanism that involved the transcription factor NF-κB. Mice deficient in miR-22, but not wild-type mice, showed attenuated T(H)17 responses and failed to develop emphysema after exposure to smoke or nCB. We further found that miR-22 controlled the activation of APCs and T(H)17 responses through the activation of AP-1 transcription factor complexes and the histone deacetylase HDAC4. Thus, miR-22 is a critical regulator of both emphysema and T(H)17 responses.


Subject(s)
Emphysema/etiology , MicroRNAs/genetics , MicroRNAs/metabolism , Repressor Proteins/antagonists & inhibitors , Th17 Cells/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Emphysema/immunology , Emphysema/metabolism , Histone Deacetylases/metabolism , Humans , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Smoking/adverse effects , Soot/toxicity , Th17 Cells/metabolism , Transcription Factor AP-1/metabolism
12.
Immunotherapy ; 7(6): 621-9, 2015.
Article in English | MEDLINE | ID: mdl-26098520

ABSTRACT

AIM: It is becoming apparent that emphysema is partly driven by self-reactive T cells inducing inflammatory damage. Thus, T cells become targets for therapy similar to other autoimmune diseases. Costimulatory blockade therapy targets disease-specific T cells, rendering them ineffective by blocking a necessary costimulatory event on the T-cell surface. This therapy is tested here in mouse emphysema. MATERIALS & METHODS: Peptides representing contact domains of counter receptors LFA-1 and ICAM-1 were used as blockade therapy in elastase-induced emphysema. RESULTS: When administered during the first week after disease induction, blockade prevented lung destruction, reduced leukocyte infiltration and inhibited the decrease in T-cell CD4:CD8 ratio, also common in human emphysema. CONCLUSION: Costimulatory blockade therapy can affect the progress of emphysema.


Subject(s)
Emphysema/therapy , Intercellular Adhesion Molecule-1/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Peptides/pharmacology , T-Lymphocytes/immunology , Animals , CD4-CD8 Ratio , Disease Models, Animal , Emphysema/immunology , Emphysema/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Peptides/immunology
13.
J Immunol ; 193(8): 3978-91, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25217166

ABSTRACT

Alpha-1 antitrypsin (AAT) deficiency (AATD) is characterized by neutrophil-driven lung destruction and early emphysema in a low AAT, and high neutrophil elastase environment in the lungs of affected individuals. In this study, we examined peripheral blood neutrophil apoptosis and showed it to be accelerated in individuals with AATD by a mechanism involving endoplasmic reticulum stress and aberrant TNF-α signaling. We reveal that neutrophil apoptosis in individuals homozygous for the Z allele (PiZZ) is increased nearly 2-fold compared with healthy controls and is associated with activation of the external death pathway. We demonstrate that in AATD, misfolded AAT protein accumulates in the endoplasmic reticulum of neutrophils, leading to endoplasmic reticulum stress and the expression of proapoptotic signals, including TNF-α, resulting in increased apoptosis and defective bacterial killing. In addition, treatment of AATD individuals with AAT augmentation therapy decreased neutrophil ADAM-17 activity and apoptosis in vivo and increased bacterial killing by treated cells. In summary, this study demonstrates that AAT can regulate neutrophil apoptosis by a previously unidentified and novel mechanism and highlights the role of AAT augmentation therapy in ameliorating inflammation in AATD.


Subject(s)
Apoptosis/immunology , Emphysema/immunology , Neutrophils/pathology , alpha 1-Antitrypsin Deficiency/drug therapy , alpha 1-Antitrypsin/therapeutic use , ADAM Proteins/biosynthesis , ADAM17 Protein , Adult , Aged , Emphysema/complications , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/immunology , Female , Humans , Inflammation/drug therapy , Inflammation/immunology , Leukocyte Elastase/biosynthesis , Leukocyte Elastase/metabolism , Lung/pathology , Lung Injury/drug therapy , Lung Injury/immunology , Lung Injury/pathology , Male , Middle Aged , Neutrophils/immunology , Protein Folding , Proteostasis Deficiencies/immunology , Pseudomonas aeruginosa/immunology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
14.
Am J Respir Crit Care Med ; 190(1): 51-61, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24874071

ABSTRACT

RATIONALE: Chronic neutrophilic inflammation is a hallmark in the pathogenesis of chronic obstructive pulmonary disease (COPD) and persists after cigarette smoking has stopped. Mechanisms involved in this ongoing inflammatory response have not been delineated. OBJECTIVES: We investigated changes to the leukotriene A4 hydrolase (LTA4H)-proline-glycine-proline (PGP) pathway and chronic inflammation in the development of COPD. METHODS: A/J mice were exposed to air or cigarette smoke for 22 weeks followed by bronchoalveolar lavage and lung and cardiac tissue analysis. Two human cohorts were used to analyze changes to the LTA4H-PGP pathway in never smokers, control smokers, COPD smokers, and COPD former smokers. PGP/AcPGP and LTA4H aminopeptidase activity were detected by mass spectroscopy, LTA4H amounts were detected by ELISA, and acrolein was detected by Western blot. MEASUREMENTS AND MAIN RESULTS: Mice exposed to cigarette smoke developed emphysema with increased PGP, neutrophilic inflammation, and selective inhibition of LTA4H aminopeptidase, which ordinarily degrades PGP. We recapitulated these findings in smokers with and without COPD. PGP and AcPGP are closely associated with cigarette smoke use. Once chronic inflammation is established, changes to LTA4H aminopeptidase remain, even in the absence of ongoing cigarette use. Acrolein modifies LTA4H and inhibits aminopeptidase activity to the same extent as cigarette smoke. CONCLUSIONS: These results demonstrate a novel pathway of aberrant regulation of PGP/AcPGP, suggesting this inflammatory pathway may be intimately involved in disease progression in the absence of ongoing cigarette smoke exposure. We highlight a mechanism by which acrolein potentiates neutrophilic inflammation through selective inhibition of LTA4H aminopeptidase activity. Clinical trial registered with www.clinicaltrials.gov (NCT 00292552).


Subject(s)
Epoxide Hydrolases/immunology , Inflammation/physiopathology , Neutrophils/immunology , Pulmonary Disease, Chronic Obstructive/etiology , Smoking/adverse effects , Aged , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/immunology , Cohort Studies , Disease Models, Animal , Emphysema/etiology , Emphysema/immunology , Female , Glycine/metabolism , Humans , Inflammation/complications , Lung/immunology , Male , Mice , Middle Aged , Myocardium/immunology , Proline/metabolism , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/physiopathology , Smoking/immunology
15.
J Immunol ; 192(11): 5354-62, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24760148

ABSTRACT

The angiopoietin-like protein 4 (angptl4, also known as peroxisome proliferator-activated receptor [PPAR]γ-induced angiopoietin-related protein) is a multifunctional protein associated with acute-phase response. The mechanisms accounting for the increase in angptl4 expression are largely unknown. This study shows that human α1-antitrypsin (A1AT) upregulates expression and release of angplt4 in human blood adherent mononuclear cells and in primary human lung microvascular endothelial cells in a concentration- and time-dependent manner. Mononuclear cells treated for 1 h with A1AT (from 0.1 to 4 mg/ml) increased mRNA of angptl4 from 2- to 174-fold, respectively, relative to controls. In endothelial cells, the maximal effect on angptl4 expression was achieved at 8 h with 2 mg/ml A1AT (11-fold induction versus controls). In 10 emphysema patients receiving A1AT therapy (Prolastin), plasma angptl4 levels were higher relative to patients without therapy (nanograms per milliliter, mean [95% confidence interval] 127.1 [99.5-154.6] versus 76.8 [54.8-98.8], respectively, p = 0.045) and correlated with A1AT levels. The effect of A1AT on angptl4 expression was significantly diminished in cells pretreated with a specific inhibitor of ERK1/2 activation (UO126), irreversible and selective PPARγ antagonist (GW9662), or genistein, a ligand for PPARγ. GW9662 did not alter the ability of A1AT to induce ERK1/2 phosphorylation, suggesting that PPARγ is a critical mediator in the A1AT-driven angptl4 expression. In contrast, the forced accumulation of HIF-1α, an upregulator of angptl4 expression, enhanced the effect of A1AT. Thus, acute-phase protein A1AT is a physiological regulator of angptl4, another acute-phase protein.


Subject(s)
Angiopoietins/immunology , Endothelial Cells/immunology , Gene Expression Regulation/immunology , Leukocytes, Mononuclear/immunology , Transcription, Genetic/immunology , alpha 1-Antitrypsin/immunology , Angiopoietin-Like Protein 1 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Anilides/pharmacology , Emphysema/drug therapy , Emphysema/immunology , Emphysema/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Gene Expression Regulation/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Male , Mitogen-Activated Protein Kinase 1/immunology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/immunology , Mitogen-Activated Protein Kinase 3/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation/drug effects , Phosphorylation/immunology , Serine Proteinase Inhibitors/immunology , Serine Proteinase Inhibitors/pharmacology , Transcription Factors/immunology , Transcription Factors/metabolism , Transcription, Genetic/drug effects , alpha 1-Antitrypsin/metabolism , alpha 1-Antitrypsin/pharmacology
16.
Inflammation ; 37(3): 745-55, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24357415

ABSTRACT

Interleukin-21 (IL-21) has been reported to be involved in many Th1-associated diseases. However, the alteration and immune regulation of IL-21 in emphysema remains unknown. In this study, we tested the levels of IFN-γ and IL-21 and the frequencies of Th1 and Tc1 in peripheral blood from cigarette smoke (CS)-exposed mice and air-exposed mice and explored the effect of IL-21 on generation of Th1 and Tc1 cells in vitro. It was found that the levels of IFN-γ and IL-21 and the frequencies of Th1, Tc1, CD4(+) IL-21(+), CD4(+) IL-21R(+), and CD8(+) IL-21R(+) T cells were much higher in CS-exposed mice. Moreover, the levels of IL-21 were correlated positively with Th1 cells and with Tc1 cells. Finally, the in vitro experiments showed that IL-21 could promote Th1/Tc1 cell generation in CS-exposed mice. These results indirectly provide evidence that IL-21 produced by CD4(+) T cells could promote Th1/Tc1 response, leading to systemic inflammation in emphysema.


Subject(s)
Emphysema/immunology , Interferon-gamma/blood , Interleukins/blood , T-Lymphocytes, Cytotoxic/immunology , Th1 Cells/immunology , Animals , Cells, Cultured , Emphysema/blood , Emphysema/chemically induced , Inflammation/immunology , Male , Mice , Mice, Inbred BALB C , Smoke/adverse effects , Smoking/adverse effects , T-Lymphocytes, Cytotoxic/cytology , Th1 Cells/cytology , Up-Regulation
17.
Eur J Pharmacol ; 721(1-3): 259-66, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-24120403

ABSTRACT

Chronic Obstructive Pulmonary Disease (COPD) is an important lung and airway disease which affects the lives of around 200 million people worldwide. The pathological hallmark of COPD is emphysema and bronchiolitis and is based on the inflammatory response of the innate and adaptive immune system to the inhalation of toxic particles and gases. The inflamed airways of COPD patients contain several inflammatory cells including neutrophils, macrophages, T lymphocytes, and dendritic cells (DC). The potential role of DCs as mediators of inflammation in the airways of smokers and COPD patients is poorly understood. The current study investigated the role of DC subsets in an animal model of cigarette smoke-induced lung emphysema through the expansion or depletion of DC subsets. Expansion of both myeloid DC (mDC) and plasmacytoid DC (pDC) by Flt3L treatment induced a decline in macrophage numbers and increased the levels of fibroblast growth factor (FGF) and vascular endothelial growth factor (VEGF) in the bronchoalveolar lavage (BAL) fluid of smoke-exposed animals. The increase in the mean linear intercept (Lm) following Flt3L treatment was decreased by pDC depletion. In conclusion, pharmacological modulation of DC subsets may have an effect on the development of airway responses and emphysema as indicated by the decline in macrophage numbers and the increase in FGF and VEGF levels in the bronchoalveolar lavage fluid. Moreover, the depletion of pDCs decreased the Lm which might suggest a role for pDC in the pathogenesis of lung emphysema.


Subject(s)
Dendritic Cells/drug effects , Emphysema/etiology , Emphysema/immunology , Smoking/adverse effects , Animals , Antibodies/metabolism , Bronchoalveolar Lavage , Dendritic Cells/immunology , Dendritic Cells/metabolism , Growth and Development/drug effects , Leukocytes/drug effects , Leukocytes/metabolism , Male , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL
18.
BMC Pulm Med ; 13: 31, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23697753

ABSTRACT

BACKGROUND: Combined pulmonary fibrosis and emphysema (CPFE) is an umbrella term encompassing upper lobe emphysema and lower lobe pulmonary fibrosis with pathogenesis elusive. The aim of our study was to investigate the incidence of autoimmune markers in patients with CPFE. METHODS: In this multicenter study we retrospectively evaluated records from patients with CPFE (n=40) and IPF (n=60) without emphysema. Baseline demographic characteristics, high-resolution computed tomography (HRCT), spirometry, histopathological, treatment, serum immunologic and survival data were investigated. B cell presence was estimated with CD20 immunostaining in representative lung biopsy samples from CPFE patients and control subjects. RESULTS: A statistically significant increased number of CPFE patients with elevated serum ANA with or without positive p-ANCA titers compared to patients with IPF without emphysema was observed. Patients with CPFE and positive autoimmune markers exhibited improved survival compared to patients with a negative autoimmune profile. A massive infiltration of clusters of CD20+ B cells forming lymphoid follicles within the fibrotic lung in CPFE patients with positive serum immunologic profile compared to patients with negative profile, was noted and positively correlated with improved survival. CONCLUSIONS: A significant proportion of patients with CPFE may present with underlying auto-immune disorders that may reside insidiously and be associated with favorable prognosis. Early identification of these patients using a panel of auto-antibodies may lead to more targeted and effective therapeutic applications.


Subject(s)
Autoimmunity/physiology , Emphysema/epidemiology , Emphysema/immunology , Pulmonary Fibrosis/epidemiology , Pulmonary Fibrosis/immunology , Adult , Aged , Antibodies, Anti-Idiotypic/blood , Antibodies, Antineutrophil Cytoplasmic/blood , Antigens, CD20/metabolism , Biomarkers/blood , Biopsy , Case-Control Studies , Comorbidity , Emphysema/mortality , Female , Humans , Kaplan-Meier Estimate , Lung/metabolism , Lung/pathology , Male , Middle Aged , Pulmonary Fibrosis/mortality , Retrospective Studies
19.
Immunol Res ; 55(1-3): 48-57, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22941590

ABSTRACT

The role of autoimmune pathology in development and progression of chronic obstructive pulmonary disease (COPD) is becoming increasingly appreciated. In this study, we identified serum autoantibody reactivities associated with chronic bronchitis or emphysema, as well as systemic autoimmunity and associated lung disease. Using autoantigen array analysis, we demonstrated that COPD patients produce autoantibodies reactive to a broad spectrum of self-antigens. Further, the level and reactivities of these antibodies, or autoantibody profile, correlated with disease phenotype. Patients with emphysema produced autoantibodies of higher titer and reactive to an increased number of array antigens. Strikingly, the autoantibody reactivities observed in emphysema were increased over those detected in rheumatoid arthritis patients, and included similar reactivities to those associated with lupus. These findings raise the possibility that autoantibody profiles may be used to determine COPD risk, as well as provide a diagnostic and prognostic tool. They shed light on the heterogeneity of autoantibody reactivities associated with COPD phenotype and could be of use in the personalization of medical treatment, including determining and monitoring therapeutic interventions.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Adult , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Bronchitis, Chronic/blood , Bronchitis, Chronic/immunology , Emphysema/blood , Emphysema/immunology , Humans , Immunoglobulin G/blood , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/immunology , Middle Aged , Phenotype , Pulmonary Disease, Chronic Obstructive/blood
20.
Am J Pathol ; 181(3): 897-906, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22901754

ABSTRACT

Transforming growth factor ß (TGF-ß) regulates inflammation, immunosuppression, and wound-healing cascades, but it remains unclear whether any of these functions involve regulation of myeloid cell function. The present study demonstrates that selective deletion of TGF-ßRII expression in myeloid phagocytes i) impairs macrophage-mediated suppressor activity, ii) increases baseline mRNA expression of proinflammatory chemokines/cytokines in the lung, and iii) enhances type 2 immunity against the hookworm parasite Nippostrongylus brasiliensis. Strikingly, TGF-ß-responsive myeloid cells promote repair of hookworm-damaged lung tissue, because LysM(Cre)TGF-ßRII(flox/flox) mice develop emphysema more rapidly than wild-type littermate controls. Emphysematous pathology in LysM(Cre)TGF-ßRII(flox/flox) mice is characterized by excessive matrix metalloprotease (MMP) activity, reduced lung elasticity, increased total lung capacity, and dysregulated respiration. Thus, TGF-ß effects on myeloid cells suppress helminth immunity as a consequence of restoring lung function after infection.


Subject(s)
Emphysema/immunology , Emphysema/pathology , Hookworm Infections/immunology , Immunity/immunology , Myeloid Cells/immunology , Nippostrongylus/immunology , Transforming Growth Factor beta/metabolism , Animals , Bone Marrow Cells/pathology , Emphysema/etiology , Emphysema/parasitology , Hookworm Infections/complications , Hookworm Infections/parasitology , Hookworm Infections/pathology , Lung/enzymology , Lung/immunology , Lung/parasitology , Lung/pathology , Lymphocyte Activation/immunology , Macrophages, Alveolar/parasitology , Macrophages, Alveolar/pathology , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia/complications , Pneumonia/immunology , Pneumonia/parasitology , Pneumonia/pathology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/metabolism , Pulmonary Fibrosis/complications , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/parasitology , Pulmonary Fibrosis/pathology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/metabolism , T-Lymphocytes/immunology , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...