Skip to main content
Erschienen in: Respiratory Research 1/2014

Open Access 01.12.2014 | Research

Combined exposure to cigarette smoke and nontypeable Haemophilus influenzae drives development of a COPD phenotype in mice

verfasst von: Shyamala Ganesan, Adam T Comstock, Brenton Kinker, Peter Mancuso, James M Beck, Uma S Sajjan

Erschienen in: Respiratory Research | Ausgabe 1/2014

Abstract

Background

Cigarette smoke (CS) is the major etiologic factor of chronic obstructive pulmonary disease (COPD). CS-exposed mice develop emphysema and mild pulmonary inflammation but no airway obstruction, which is also a prominent feature of COPD. Therefore, CS may interact with other factors, particularly respiratory infections, in the pathogenesis of airway remodeling in COPD.

Methods

C57BL/6 mice were exposed to CS for 2 h a day, 5 days a week for 8 weeks. Mice were also exposed to heat-killed non-typeable H. influenzae (HK-NTHi) on days 7 and 21. One day after the last exposure to CS, mice were sacrificed and lung inflammation and mechanics, emphysematous changes, and goblet cell metaplasia were assessed. Mice exposed to CS or HK-NTHi alone or room air served as controls. To determine the susceptibility to viral infections, we also challenged these mice with rhinovirus (RV).

Results

Unlike mice exposed to CS or HK-NTHi alone, animals exposed to CS/HK-NTHi developed emphysema, lung inflammation and goblet cell metaplasia in both large and small airways. CS/HK-NTHi-exposed mice also expressed increased levels of mucin genes and cytokines compared to mice in other groups. CS/HK-NTHi-exposed mice infected with RV demonstrated increased viral persistence, sustained neutrophilia, and further increments in mucin gene and chemokine expression compared to other groups.

Conclusions

These findings indicate that in addition to CS, bacteria may also contribute to development of COPD, particularly changes in airways. Mice exposed to CS/HK-NTHi are also more susceptible to subsequent viral infection than mice exposed to either CS or HK-NTHi alone.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1465-9921-15-11) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SG, designed and conducted the experiments and analyzed the data; ATC and BK, provided technical support; PM and JMB, contributed intellectually; US, conceived and designed the study and wrote the manuscript. All authors read and approved the final manuscript.
Abkürzungen
COPD
Chronic obstructive pulmonary disease
RV
Rhinovirus
HK-NTHi
Heat killed nontypeable Haemophilus influenzae
CS
Cigarette smoke
RA
Room air
P-V
Pressure - volume.

Background

Chronic obstructive pulmonary disease (COPD) is the fourth leading cause of death world-wide, and the prevalence of COPD is increasing globally [1]. COPD is characterized by airflow limitation that is progressive and is usually irreversible. Small airway remodeling, including narrowing of airways due to peribronchiolar fibrosis, goblet cell metaplasia and excessive mucus production, is now accepted as an important cause of airflow obstruction in COPD [2]. In addition, lung parenchyma is destroyed by proteolytic damage (emphysema), reducing the elasticity and gas-exchange surface area of the lung. Acute exacerbations further enhance airways obstruction and accelerate progression of lung disease in these patients (reviewed in [3]). However, the underlying mechanisms for these changes are not well understood, partly because small-animal models do not recapitulate all the typical features of human COPD.
COPD has been modeled in mice by administration of proteases, lipopolysaccharide (LPS), chemicals and cigarette smoke (CS). In particular, CS has been used extensively to investigate mechanisms of COPD pathogenesis since it is the major risk factor in the development of COPD [3]. While whole body exposure to CS for a short-term (3 days to 4 weeks) has been useful in evaluating the mechanisms of CS-induced acute lung inflammation and defective innate immune responses to subsequent infections [47], long-term exposure to CS for periods up to 6 months has been employed to understand the mechanisms of emphysema development [811]. However, neither of these models show changes in small airways, which plays a major role in the development of airflow limitation in COPD implying that other factors in addition to CS are required to mimic COPD lung disease in mice [12].
Although CS exposure is the key insult in the pathogenesis of COPD, only 25 to 35% of smokers develop COPD [13, 14] suggesting the contribution of other factors, such as genetic background, concurrent respiratory infections, and aberrant host responses in the development of COPD. Respiratory pathogens including bacteria, viruses and fungi are often present in the airways of COPD patients and therefore it is plausible that respiratory pathogens or their products such as enterotoxin, endotoxin, viral RNA may contribute to disease pathogenesis [1517]. For example Kang et al. demonstrated that CS synergizes with synthetic double stranded (ds) RNA, a viral RNA mimetic to induce enhanced inflammatory and emphysematous changes, and airway fibrosis in the mouse lungs [17]. However these mice did not develop goblet cell metaplasia, increased mucus production or airways obstruction, which are also important features of COPD. Similar results were observed when mice were infected with influenza virus instead of treating with dsRNA. Enterotoxin B isolated from S. aureus was shown to exacerbate CS-induced inflammatory changes in mouse lungs and induce goblet cell metaplasia and formation of lymphoid aggregates, but these mice did not develop emphysema. Chronic exposure of mice to lysates of non-typeable H. influenzae (NTHi) was shown to induce airway inflammation but not emphysema or airway remodeling [18]. Endotoxin, a bacterial cell wall component is present in abundant amounts as a contaminant in CS [19, 20] and prolonged intratracheal exposure of mice to endotoxin, (twice week for 3 months) induces lung inflammation, and changes in both parenchyma and airways which persists up to 8 weeks [21]. We demonstrated that exposure of mice once a week to combination of elastase and endotoxin for 4 weeks induces all the features of COPD [22]. The latter three models although exhibit features of COPD and indicate involvement of bacterial factors in COPD pathogenesis, these models may not be representative of CS-induced changes. Based on these observations, in the present study we examine a novel concept that in addition to CS, exposure to bacteria is required to induce typical features of COPD including emphysema, airway remodeling, and lung inflammation in mice. Since NTHi is frequently isolated from clinically stable COPD patients as well as during exacerbations [23], we used NTHi in the present study. We also evaluated susceptibility to rhinovirus (RV) infection in this model, because RV is associated with virally mediated exacerbations of COPD and sometimes leads to progression of lung disease [24, 25].

Methods

Animals and treatment

Eight to ten week old C57BL/6 mice (Charles River Laboratories, Wilmington, MA) were exposed to CS as described previously [26]. Briefly mice were exposed to smoke from standardized 3R4F research cigarettes (University of Kentucky, Lexington, KY) generated by a TE-2 cigarette smoking machine (Teague Enterprises, Woodland, CA). This device is set up to provide a mixture of mainstream and sidestream smoke. Animals were exposed to CS for 2 h/day, 5 days/week for 8 consecutive weeks in a 54-L glass and Plexiglas whole-body exposure chamber with an electric fan for chamber mixing in standard mouse caging units with wire cage tops, with water available ad libitum. The mean concentration of particulates collected during a 2-h exposure was 9.28 ± 1.45 mg/day. Control mice were exposed to room air (RA) under similar conditions. Heat-killed (HK) NTHi was administered into the lungs on days 7 and 21 by intranasal route. Three days after the last exposure to CS, lungs were lavaged [27] to determine total and differential cell counts and to measure cytokines, were fixed in formalin and embedded in paraffin [22] to evaluate morphology, or were homogenized in PBS to isolate total RNA. In additional experiments, mice were inoculated intranasally with RV or with a sham preparation, and sacrificed 4 days post infection. All experiments were approved by the Animal Care and Use Committee of the University of Michigan and Veterans Affairs, Ann Arbor Medical Center Animal Care Committee.

Heat killed bacteria

NTHi strain, 5P54H1 was isolated from a COPD patient and was kindly provided by Dr. Timothy Murphy, University of Buffalo. Bacteria were cultured on chocolate agar, suspended in PBS to density of 1 × 109 CFU/ml and incubated at 80°C for 30 min to kill the bacteria. Mice received 50 μl of heat-killed (HK) NTHi or PBS intranasally [28].

Rhinovirus and infection

Stocks of RV1B were prepared by infecting HeLa cells with RV1B and subjecting HeLa cell supernatants to ultrafiltration and then viral titer was determined as described previously [22, 29]. Similarly concentrated and purified cell supernatants from uninfected HeLa cells were used as sham controls. At the end of 8 weeks of exposure to CS or RA and +/- HK-NTHi, mice were infected with RV or an equal volume of sham preparation by intranasal route as described previously [22] and sacrificed 4 days post infection.

Bronchoalveolar Lavage (BAL)

After relevant treatment, mice were sacrificed and BAL was performed by instilling 1 ml PBS containing 5 mM EDTA 10 times [27]. The first wash was centrifuged and supernatant was retained for determination of cytokines by ELISA. Cells from the first wash were combined with the cells from rest of the washes and suspended in 1 ml of PBS and numbers of cells were counted to determine total cells. Cytospins prepared from BAL cells were stained with Diff-Quick and differential counts were determined by counting a minimum of 200 cells.

RNA isolation and qPCR

Lungs were collected under aseptic conditions after relevant treatment and homogenized in 2 ml PBS. An aliquot of lung homogenate was immediately mixed with TRIZOL and total RNA was then purified by using RNeasy miRNA kit (Qiagen, Alameda, CA). Total RNA was reverse transcribed to first strand cDNA using Taqman reverse transcription kit (Applied Biosystems Life Technologies, Carlsbad, CA). cDNA was then used to determine the mRNA expression of IP-10, Muc5B, Muc5AC, Gob5 and G3PDH (house-keeping gene) by quantitative qPCR using gene specific primers and Syber green PCR mix. All primers were designed and purchased from IDT (Coralville, IA). To detect viral RNA, total RNA isolated from the lungs was subjected quantitative Taqman qPCR as described previously [30] and expressed as number of vRNA copies per 10 μg of total RNA.

Infectious viral load

Lung homogenates were used to determine the infectious viral load by plaque assay as described [22].

Histology and morphometry

Lungs were inflation fixed at a constant pressure of 30 cm.H2O for 30 min. Lungs were embedded in paraffin, and 5 μ thick sagittal sections were stained with hematoxylin and eosin (H & E) to assess histology or periodic acid-Schiff (PAS) to detect goblet cells. Alveolar chord length was determined as described previously [22, 31]. Briefly, sagittal sections obtained at 5 mm intervals through the depth of the lungs were stained with H & E and the diameters of the air spaces measured in at least 10 random areas using NIH image J analysis software. Sagittal sections obtained at 5 mm intervals were also stained with PAS and the number of PAS positive cells per 100 μM basement membrane lengths was counted in random fields. Airways with perimeter of ≤1 mM were considered as small airways. Airways with cartilage were considered as large airways [22].

Lung mechanics and airways resistance

Dynamic lung elastance and compliance, pressure-volume relationship in mice exposed to RA, CS, RA/HK-NTHi or CS/HK-NTHi, were measured as described previously using a miniature computerized flexivent ventilator (Scireq, Canada) [22]. Airways responsiveness to increasing dose of methacholine challenge following RV infection was measured by plethysmography [30].

ELISA

Bronchoalveolar lavage fluid was centrifuged and the supernatant was used for assessing the levels of chemokines and cytokines by ELISA.

Statistical Analysis

Results are expressed as means ± SD or mean with range of data. Data were analyzed by using SigmaStat statistical software (Systat Software, San Jose, CA). One- or two-way ANOVA with Tukey’s range test, ANOVA on ranks with Friedman’s test or unpaired t test was performed as appropriate to compare groups and a P value ≤ 0.05 was considered significant.

Results

Lung histology

Lung sections from mice exposed to room air (RA), RA/HK-NTHi, CS, CS-HK-NTHi were evaluated by light microscopy. RA-exposed mice showed normal morphology with baseline air space size and no appreciable inflammation (Figure 1A and 1B). CS-exposed mice showed mild lung inflammation, enlarged airspaces in a patchy distribution, and accumulated inflammatory cells (possibly macrophages) in the airspaces (Figure 1C and 1D). Mice exposed to HK-NTHi showed no enlarged airspaces, but demonstrated peribronchiolar inflammation (Figure 1E to 1H). Mice treated with a combination of CS and HK-NTHi showed enlarged airspaces and mild lung inflammation (Figure 1I), as well as accumulation of inflammatory cells in peribronchiolar and perivascular areas (Figure 1J and 1K) and in the airspaces (Figure 1L). Compared to RA- or CS-exposed mice, both HK-NTHi and CS/HK-NTHi-exposed mice showed increases in the numbers of PAS positive cells (indicative of goblet cell metaplasia) in large airway epithelia, however it was more pronounced in CS/NK-NTHi-exposed mice (Figure 2A to 2D). Mice in CS/HK-NTHi group, but not animals in other groups showed increased numbers of PAS positive cells in small airway epithelia (Figure 2E to 2H). Mice exposed to HK-NTHi or CS/HK-NTHi also showed airway epithelial hyperplasia in both small and large airways, but it was more prominent in CS/HK-NTHi-exposed mice.

Morphometry and lung mechanics

Morphometric analysis was performed to determine the mean airspace chord length. Compared to RA-exposed mice both CS- and CS/HK-NTHi-exposed mice showed small but significant increases in chord length (RA vs CS, p = 0.045; RA vs CS/HK-NTHi, p = 0.002) (Figure 3A), but mice exposed to HK-NTHi did not. The changes in chord length in mice exposed to combination of CS and HK-NTHi were higher than in mice exposed to CS alone (p = 0.031).
We assessed elasticity of the lungs by determining total respiratory system compliance, elastance, and the pressure-volume (P-V) relationships during inflation. Compared to RA- or HK-NTHi-exposed mice, CS and CS/HK-NTHi- exposed mice showed increased compliance (CS vs RA, p = 0.047; CS vs HK-NTHi, p = 0.039; CS/HK-NTHi vs RA, p = 0.022; CS/HK-NTHi vs HK-NTHi, p = 0.029) (Figure 3B). Conversely, a significant decreases in elastance (Figure 3C) were observed in CS or CS/HK-NTHi group compared to mice exposed to RA or HK-NTHi (CS vs RA, p = 0.038; CS vs HK-NTHi, p = 0.025, CS/HK-NTHi vs RA, p = 0.011; CS/HK-NTHi vs HK-NTHi, p = 0.007). However, the changes in both compliance and elastance were significantly higher in CS/HK-NTHi-exposed mice than in CS-exposed mice (compliance, p = 0.034 and elastance, p = 0.021). Consistent with these observations, we also observed slightly upward and leftward shifts in the P-V relationships in CS- and CS/HK-NTHi-exposed mice compared to mice in other two groups, demonstrative of reduced elastic recoil. However, the changes in P-V relationship were more pronounced in CS/HK-NTHi-exposed mice (Figure 3D).

Airways inflammation and mucin gene expression

To assess lung inflammation, we performed total and differential cell counts and quantified levels of IL-1β, TNF-α, KC and MCP-1 by ELISA. Compared to mice exposed to RA or CS, mice in HK-NTHi and CS/HK-NTHi groups showed a significantly higher numbers of total cells (RA vs HK-NTHi, p = 0.043; CS vs HK-NTHi, p = 0.048; RA vs CS/HK-NTHi p = 0.013; CS vs CS/HK-NTHi, p = 0.025) with the largest increase observed in the CS/HK-NTHi group (HK-NTHi vs CS/HK-NTHi, p = 0.0103) (Figure 4A). Both macrophages (Figure 4B) and lymphocytes were higher (Figure 4C) in the airways of CS/HK-NTHi than in RA-exposed mice (p = 0.027 and p = 0.031 respectively). Only the number of lymphocytes increased significantly in CS, HK-NTHi-exposed mice compared to RA-exposed mice (p = 0.049 and p = 0.043 respectively). There were no significant differences in numbers of neutrophils among the groups (data not shown). We then examined the levels of cytokines in the lavage fluid (Figure 4D to 4G). Compared to mice exposed to RA, mice in all other groups showed more IL-1β (RA vs CS, p = 0.043; RA vs HK-NTHi, p = 0.042; RA vs CS/HK-NTHi, p = 0.037) and KC levels (RA vs CS, p = 0.010; RA vs HK-NTHi, p = 0.007; RA vs CS/HK-NTHi, p = 0.023). Mice in HK-NTHi or CS/HK-NTHi groups, but not in the CS group showed higher levels of MCP-1 (RA vs HK-NTHi, p = 0.042; RA vs CS/HK-NTHi, p = 0.001) and TNF-α (RA vs HK-NTHi, p = 0.049; RA vs CS/HK-NTHi, p = 0.02) than the RA group. However, mice in the CS/HK-NTHi group showed the highest increases in the levels of all the cytokines measured.
We then measured the expression of mucin genes, Muc5B and Muc5AC and a calcium-activated chloride channel that is thought to regulate mucus production and/or secretion, Gob5 (mclca3) [32] by qPCR. The expression of Muc5AC and Gob5, but not Muc5B was increased in HK-NTHi-exposed mice compared to RA-exposed, but not in CS-exposed mice (Muc5AC- RA vs HK-NTHi, p = 0.041; Gob5- RA vs HK-NTHi, p = 0.038) (Figure 5A to 5C). In contrast, CS/HK-NTHi-exposed mice showed increases in Muc5B (p = 0.041) and Muc5AC (p = 0.045) as well as Gob5 (p = 0.039) compared to RA-exposed mice. Again CS/HK-NTHi mice showed the largest increases in expression in all three genes.

Susceptibility to RV infection

At the end of 8 weeks of exposure to CS, mice were infected with RV or sham, sacrificed after 4 days and examined for viral RNA, chemokine expression, neutrophil infiltration and mucin gene expression. CS/HK-NTHi-exposed mice infected with RV showed one log higher vRNA than similarly infected animals in other groups (p = 0.024) (Figure 6A). CS/HK-NTHi mice also showed higher infectious viral load than mice in other groups (p = 0.007), but it was only 1 to 2 × 102 PFU/lung (Figure 6B). Despite having low levels of infectious viral load, the CS/HK-NTHi group showed remarkably higher neutrophil (p = 0.001) and lymphocyte (p = 0.009) infiltration compared to the sham infected group (Figure 6C and 6D). Consistent with the cellular infiltration, CS/HK-NTHi mice infected with RV also showed persistent increases in KC (p = <0.001), MIP-2 (p = 0.001) and IP-10 (0.022), compared to sham infected mice (Figure 6E-6G). RV infected mice in other groups showed very small increases in infiltrated neutorphils, lymphocytes and cytokine levels over their respective sham-infected controls (p values ranged between 0.039 to 0.05 in these animals).
To assess the effects of RV infection on airways obstruction, we examined the expression of mucin genes, Gob5, goblet cell metaplasia in small airways, and responses to methacholine challenge. Irrespective of infection, mice exposed to RA, CS or RA/HK-NTHi did not show changes in the expression of Muc5B, Muc5AC or Gob5 (data not shown). In contrast, CS/HK-NTHi-exposed mice infected with RV showed additional increases in the expression of Muc5AC (p = 0.041) and Gob5 (p = 0.003), but not muc2B compared to similarly-exposed sham-infected mice (Figure 7A to 7C). CS/HK-NTHi-exposed mice infected with RV also showed a further increment in PAS-positive cells in the small airways compared to sham-infected mice, implying that enhanced mucus expression may be due to increased goblet cell metaplasia in these mice (Figure 7D and 7E). In contrast, mice exposed to RA, CS or RA/HK-NTHi infected with RV did not show an increase in either the expression of mucin genes or the number of goblet cells in the airway epithelium (data not shown).
RV-infected mice, irrespective of prior exposure to CS. NTHi, or combination of CS/NTHi, showed an increase in airways resistance in response to methacholine challenge over respective sham-infected mice (Figure 8A to 8D). However, CS/HK-NTHi-exposed mice infected with RV showed a much higher increase in airways resistance to methacholine challenge than similarly infected mice in other groups.
Taken together these results demonstrate that in addition to CS, bacterial products may also be required to induce COPD-like changes in both parenchyma and conducting airways of mice, and that CS/HK-NTHi-exposed mice are also more susceptible to subsequent RV infection.

Discussion

Exacerbations in COPD patients are characterized by increased inflammation and increased lower respiratory tract symptoms requiring change in therapy such as treatment with antibiotics, steroids or antiviral drugs [33]. Exacerbations may be triggered primarily by viral or bacterial infections and are often associated with accelerated progression of lung disease. RV, which causes self-limiting infections in healthy individuals is responsible for the majority of virus-related exacerbations in patients with COPD [24, 25], but the underlying mechanisms are not well understood. In this study, we developed a mouse model which displays phenotypic characteristics of COPD, including emphysema, mild but diffuse lung inflammation, and goblet cell metaplasia. We combined this model with our previously described mouse model of RV infection [22, 34] and show that these mice show prolonged neutrophilic lung inflammation and airways obstruction, similar to that observed in mild COPD patients experimentally infected with RV [35], indicating the suitability of this model to elucidate mechanisms of COPD exacerbations.
The mouse model of COPD developed in this study supports our primary hypothesis that both CS and bacteria are required for development of COPD–like changes in mice and also render mice susceptible to viral infection. We demonstrate that although CS is the major risk factor in the development of COPD, mice exposed to CS alone for 8 weeks only develop mild lung inflammation and emphysema but not small airway disease which is consistent with previous observations [4, 811]. In contrast, exposure of mice to a combination of CS and NTHi induces not only more pronounced emphysema and lung inflammation than mice exposed to CS alone, but also goblet cell metaplasia in the airways, which is one of the pathologic features of COPD. In addition, mice exposed to combination of CS/HK-NTHi also show heightened susceptibility to viral infection with further increases in lung inflammation and goblet cell metaplasia. Together, these findings suggest that this novel mouse model of COPD is not only useful in understanding the mechanisms of exacerbations, but also for delineating the mechanisms underlying progression of lung disease in COPD. However, it should be noted that continuous exposure to CS and/or intermittent infection with bacteria or virus may be required to maintain chronicity of the COPD-like changes in these mice.
NTHi is one of the more commonly isolated organisms from clinically stable COPD patients and also at exacerbations [23], and patients who are chronically colonized with NTHi show significantly more neutrophilic airway inflammation than those who are not colonized [15, 36]. Further, repeated exposure to extracts of heat-killed NTHi causes airway inflammation in mice with cellular and cytokine profiles similar to COPD [18]. Finally, NTHi products increase mucin gene expression in vivo and in vitro [37]. These observations indicate that NTHi may contribute to the development of COPD particularly, small airway disease and lung inflammation. Based on these facts, we postulated that NTHi may synergize with CS to induce development of COPD-like features in mice which includes changes in both small airways and parenchyma. In the present study, we opted to use low-dose (5 × 106 CFU) heat-killed NTHi to induce milder and sustained inflammation with minimal infiltration of neutrophils as opposed to acute neutrophil-dominated inflammation induced by live NTHi [27, 28], to prevent extensive lung damage. The bacterial dose was chosen based on our preliminary experiments, in which mice were treated with heat-killed bacteria equivalent to 5 × 105, 5 × 106 or 5 × 107 CFU once a week for 4 consecutive weeks and sacrificed 4 weeks after the last treatment. By morphology, mice treated with 5 × 105 CFU did not show any detectable changes in the lungs and were very similar to untreated mice. In contrast, mice treated with 5 × 107 CFU showed severe lung inflammation, consolidation of parenchyma and pronounced goblet cell metaplasia in both small and large airways. Mice treated with 5 × 106 CFU showed mild to moderate lung inflammation with mild goblet cell metaplasia. In addition, we also found that two exposures to 5 × 106 CFU instead of four exposures were sufficient to induce these changes in the lung. Combination of this HK-NTHi treatment with CS however, led to more pronounced lung inflammation and goblet cell metaplasia, increased mucin gene expression and also the development of emphysema. Interestingly these mice also showed thickening of airway epithelia, similar to the airway epithelial hyperplasia observed in COPD patients [38]. We also observed aggregates of inflammatory cells particularly in the peribronchiolar and perivascular areas resembling lymphoid aggregates [16], but the nature of these aggregates is yet to be determined. We speculate that these pronounced pathological changes in CS/HK-NTHi-exposed mice may be the result of exaggerated host innate immune responses to bacterial products, in the presence of CS.
Alveolar macrophages in the lungs play an important role in clearing bacteria and limit bacteria-induced inflammation. CS has been demonstrated to affect the function of alveolar macrophages by shifting their phenotype from M2 to M1 [39, 40]. M1 macrophages respond to bacterial antigens such as LPS by producing relatively more inflammatory cytokines than M2 macrophages [41]. Although this is required for clearance of bacteria, sustained production of inflammatory cytokines may increase lung inflammation, emphysema and also induce goblet cell metaplasia [8, 42]. Based on these observations, we speculate that CS-induced shift in macrophage phenotype may be one of the mechanisms by which CS synergizes with NTHi to increase expression of inflammatory cytokines and this in turn may lead to increased lung inflammation, emphysema and goblet cell metaplasia. Bacterial pathogens or their products other than NTHi may also induce COPD-like changes when combined with CS, but the magnitude of pathological changes may vary depending on the bacteria used. However, further studies are required to confirm this notion.
CS/HK-NTHi-exposed mice displaying the COPD phenotype were also found to be susceptible to subsequent infection with RV, a virus associated with a majority of viral-associated COPD exacerbations [35, 43]. Four days after challenging with RV, CS/HK-NTHi-exposed mice despite showing minimally higher viral loads than similarly infected RA-, CS-, or NTHi-exposed mice, displayed sustained neutrophilia and lymphocyte infiltration, and increased expression of KC, MIP-2 and IP-10, which was not observed in mice from other groups. These changes were associated with progression in goblet cell metaplasia in small airways and increased expression of Muc5AC (mucin gene) and Gob5, a chloride channel that plays a role in mucin secretion [32]. These mice also showed sustained airways hyperresponsiveness to methacholine challenge. Previously, we have shown that airway epithelial cells isolated from COPD patients are more susceptible to RV infection despite expressing interferon and other antiviral genes [44]. This is probably due to increase in the number of goblet cells in COPD cell cultures, because previously goblet cells have been shown to be more permissive for RV infection [45]. Therefore it is plausible that CS/HK-NTHi mice which show increased numbers of goblet cells in their airways may be more susceptible to RV infection. In addition, exposure to CS may also support replication of virus and induce aberrant production of inflammatory cytokines. Consistent with this notion, acute exposure to CS was shown to increase production of pro-inflammatory cytokines and reduce expression of antiviral genes in response to RV infection in airway epithelial cells [46, 47]. Such aberrant increases in inflammatory cytokines following RV infection may further enhance lung inflammation, goblet cell metaplasia and airways obstruction. However further studies are required to confirm this notion. These observations are consistent with human studies, in which mild COPD patients were shown to develop sustained lower respiratory tract symptoms and increased infiltration of neutrophil and lymphocyte in the lungs after experimental rhinovirus infection [35]. These changes correlated with decreases in lung function in these patients. Another noteworthy observation is that CS/HK-NTHi-exposed mice infected with RV also showed sustained increases in IP-10 levels, which has been proposed to be one of the biomarkers of COPD exacerbations [48, 49]. These similarities in responses to RV infection between COPD patients and CS/HK-NTHi mice imply that this model may be useful in understanding the molecular mechanisms related to viral-associated COPD exacerbations and progression of lung disease.

Conclusions

In summary, we show that CS synergizes with NTHi to cause a COPD-like phenotype in mice within a short period of time. As far as we know, this is the first mouse model of COPD to display innate immune responses to RV infection similar to that observed in COPD patients. Therefore we believe that this model could be used to elucidate the mechanisms of disease progression following acute exacerbations particularly that are associated with viral infections. This model may also be useful to model a chronic state of COPD, provided continuation of exposure to CS in addition to intermittent administration of bacterial or viral products.

Acknowledgments

Supported by NIH grants AT004793 (US), HL089772 (US), and FAMRI grant CIA-103071(PM) and Research Funds from the Department of Medicine, University of Michigan (JMB).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver ( https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​ ) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

SG, designed and conducted the experiments and analyzed the data; ATC and BK, provided technical support; PM and JMB, contributed intellectually; US, conceived and designed the study and wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Kochanek KD, Xu J, Murphy SL, Minono AM, Kung H: Deaths:Preliminary Data for 2009. National Vitral Statistics Reports. 2011, 59: 1-51. Kochanek KD, Xu J, Murphy SL, Minono AM, Kung H: Deaths:Preliminary Data for 2009. National Vitral Statistics Reports. 2011, 59: 1-51.
2.
Zurück zum Zitat Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD: The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004, 350: 2645-2653. 10.1056/NEJMoa032158.PubMedCrossRef Hogg JC, Chu F, Utokaparch S, Woods R, Elliott WM, Buzatu L, Cherniack RM, Rogers RM, Sciurba FC, Coxson HO, Pare PD: The nature of small-airway obstruction in chronic obstructive pulmonary disease. N Engl J Med. 2004, 350: 2645-2653. 10.1056/NEJMoa032158.PubMedCrossRef
3.
Zurück zum Zitat Mannino DM, Buist AS: Global burden of COPD: risk factors, prevalence, and future trends. Lancet. 2007, 370: 765-773. 10.1016/S0140-6736(07)61380-4.PubMedCrossRef Mannino DM, Buist AS: Global burden of COPD: risk factors, prevalence, and future trends. Lancet. 2007, 370: 765-773. 10.1016/S0140-6736(07)61380-4.PubMedCrossRef
4.
Zurück zum Zitat Gaschler GJ, Skrtic M, Zavitz CC, Lindahl M, Onnervik PO, Murphy TF, Sethi S, Stampfli MR: Bacteria challenge in smoke-exposed mice exacerbates inflammation and skews the inflammatory profile. Am J Respir Crit Care Med. 2009, 179: 666-675. 10.1164/rccm.200808-1306OC.PubMedCrossRef Gaschler GJ, Skrtic M, Zavitz CC, Lindahl M, Onnervik PO, Murphy TF, Sethi S, Stampfli MR: Bacteria challenge in smoke-exposed mice exacerbates inflammation and skews the inflammatory profile. Am J Respir Crit Care Med. 2009, 179: 666-675. 10.1164/rccm.200808-1306OC.PubMedCrossRef
5.
Zurück zum Zitat Doz E, Noulin N, Boichot E, Guenon I, Fick L, Le Bert M, Lagente V, Ryffel B, Schnyder B, Quesniaux VF, Couillin I: Cigarette smoke-induced pulmonary inflammation is TLR4/MyD88 and IL-1R1/MyD88 signaling dependent. J Immunol. 2008, 180: 1169-1178.PubMedCrossRef Doz E, Noulin N, Boichot E, Guenon I, Fick L, Le Bert M, Lagente V, Ryffel B, Schnyder B, Quesniaux VF, Couillin I: Cigarette smoke-induced pulmonary inflammation is TLR4/MyD88 and IL-1R1/MyD88 signaling dependent. J Immunol. 2008, 180: 1169-1178.PubMedCrossRef
6.
Zurück zum Zitat Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J, Shapiro SD, Wright JL: Macrophage metalloelastase mediates acute cigarette smoke-induced inflammation via tumor necrosis factor-alpha release. Am J Respir Crit Care Med. 2003, 167: 1083-1089. 10.1164/rccm.200212-1396OC.PubMedCrossRef Churg A, Wang RD, Tai H, Wang X, Xie C, Dai J, Shapiro SD, Wright JL: Macrophage metalloelastase mediates acute cigarette smoke-induced inflammation via tumor necrosis factor-alpha release. Am J Respir Crit Care Med. 2003, 167: 1083-1089. 10.1164/rccm.200212-1396OC.PubMedCrossRef
7.
Zurück zum Zitat Onnervik PO, Lindahl M, Svitacheva N, Stampfli M, Thim K, Smailagic A, Virtala R, Taylor JD: The role of the CCR1 receptor in the inflammatory response to tobacco smoke in a mouse model. Inflamm Res. 2010, 59: 817-825. 10.1007/s00011-010-0193-5.PubMedPubMedCentralCrossRef Onnervik PO, Lindahl M, Svitacheva N, Stampfli M, Thim K, Smailagic A, Virtala R, Taylor JD: The role of the CCR1 receptor in the inflammatory response to tobacco smoke in a mouse model. Inflamm Res. 2010, 59: 817-825. 10.1007/s00011-010-0193-5.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Churg A, Wang RD, Tai H, Wang X, Xie C, Wright JL: Tumor necrosis factor-alpha drives 70% of cigarette smoke-induced emphysema in the mouse. Am J Respir Crit Care Med. 2004, 170: 492-498. 10.1164/rccm.200404-511OC.PubMedCrossRef Churg A, Wang RD, Tai H, Wang X, Xie C, Wright JL: Tumor necrosis factor-alpha drives 70% of cigarette smoke-induced emphysema in the mouse. Am J Respir Crit Care Med. 2004, 170: 492-498. 10.1164/rccm.200404-511OC.PubMedCrossRef
9.
Zurück zum Zitat Rinaldi M, Maes K, De Vleeschauwer S, Thomas D, Verbeken EK, Decramer M, Janssens W, Gayan-Ramirez GN: Long-term nose-only cigarette smoke exposure induces emphysema and mild skeletal muscle dysfunction in mice. Dis Model Mech. 2012, 5: 333-341. 10.1242/dmm.008508.PubMedPubMedCentralCrossRef Rinaldi M, Maes K, De Vleeschauwer S, Thomas D, Verbeken EK, Decramer M, Janssens W, Gayan-Ramirez GN: Long-term nose-only cigarette smoke exposure induces emphysema and mild skeletal muscle dysfunction in mice. Dis Model Mech. 2012, 5: 333-341. 10.1242/dmm.008508.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Yao H, Chung S, Hwang JW, Rajendrasozhan S, Sundar IK, Dean DA, McBurney MW, Guarente L, Gu W, Ronty M, et al: SIRT1 protects against emphysema via FOXO3-mediated reduction of premature senescence in mice. J Clin Invest. 2012, 122: 2032-2045. 10.1172/JCI60132.PubMedPubMedCentralCrossRef Yao H, Chung S, Hwang JW, Rajendrasozhan S, Sundar IK, Dean DA, McBurney MW, Guarente L, Gu W, Ronty M, et al: SIRT1 protects against emphysema via FOXO3-mediated reduction of premature senescence in mice. J Clin Invest. 2012, 122: 2032-2045. 10.1172/JCI60132.PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat D’Hulst AI, Vermaelen KY, Brusselle GG, Joos GF, Pauwels RA: Time course of cigarette smoke-induced pulmonary inflammation in mice. Eur Respir J. 2005, 26: 204-213. 10.1183/09031936.05.00095204.PubMedCrossRef D’Hulst AI, Vermaelen KY, Brusselle GG, Joos GF, Pauwels RA: Time course of cigarette smoke-induced pulmonary inflammation in mice. Eur Respir J. 2005, 26: 204-213. 10.1183/09031936.05.00095204.PubMedCrossRef
12.
Zurück zum Zitat Makita H, Nasuhara Y, Nagai K, Ito Y, Hasegawa M, Betsuyaku T, Onodera Y, Hizawa N, Nishimura M: Characterisation of phenotypes based on severity of emphysema in chronic obstructive pulmonary disease. Thorax. 2007, 62: 932-937. 10.1136/thx.2006.072777.PubMedPubMedCentralCrossRef Makita H, Nasuhara Y, Nagai K, Ito Y, Hasegawa M, Betsuyaku T, Onodera Y, Hizawa N, Nishimura M: Characterisation of phenotypes based on severity of emphysema in chronic obstructive pulmonary disease. Thorax. 2007, 62: 932-937. 10.1136/thx.2006.072777.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Lokke A, Lange P, Scharling H, Fabricius P, Vestbo J: Developing COPD: a 25 year follow up study of the general population. Thorax. 2006, 61: 935-939. 10.1136/thx.2006.062802.PubMedPubMedCentralCrossRef Lokke A, Lange P, Scharling H, Fabricius P, Vestbo J: Developing COPD: a 25 year follow up study of the general population. Thorax. 2006, 61: 935-939. 10.1136/thx.2006.062802.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Ohar JA, Sadeghnejad A, Meyers DA, Donohue JF, Bleecker ER: Do symptoms predict COPD in smokers?. Chest. 2010, 137: 1345-1353. 10.1378/chest.09-2681.PubMedPubMedCentralCrossRef Ohar JA, Sadeghnejad A, Meyers DA, Donohue JF, Bleecker ER: Do symptoms predict COPD in smokers?. Chest. 2010, 137: 1345-1353. 10.1378/chest.09-2681.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Sethi S, Murphy TF: Infection in the pathogenesis and course of chronic obstructive pulmonary disease. N Engl J Med. 2008, 359: 2355-2365. 10.1056/NEJMra0800353.PubMedCrossRef Sethi S, Murphy TF: Infection in the pathogenesis and course of chronic obstructive pulmonary disease. N Engl J Med. 2008, 359: 2355-2365. 10.1056/NEJMra0800353.PubMedCrossRef
16.
Zurück zum Zitat Huvenne W, Lanckacker EA, Krysko O, Bracke KR, Demoor T, Hellings PW, Brusselle GG, Joos GF, Bachert C, Maes T: Exacerbation of cigarette smoke-induced pulmonary inflammation by Staphylococcus aureus enterotoxin B in mice. Respir Res. 2011, 12: 69-PubMedPubMedCentralCrossRef Huvenne W, Lanckacker EA, Krysko O, Bracke KR, Demoor T, Hellings PW, Brusselle GG, Joos GF, Bachert C, Maes T: Exacerbation of cigarette smoke-induced pulmonary inflammation by Staphylococcus aureus enterotoxin B in mice. Respir Res. 2011, 12: 69-PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Kang MJ, Lee CG, Lee JY, Dela Cruz CS, Chen ZJ, Enelow R, Elias JA: Cigarette smoke selectively enhances viral PAMP- and virus-induced pulmonary innate immune and remodeling responses in mice. J Clin Invest. 2008, 118: 2771-2784.PubMedPubMedCentral Kang MJ, Lee CG, Lee JY, Dela Cruz CS, Chen ZJ, Enelow R, Elias JA: Cigarette smoke selectively enhances viral PAMP- and virus-induced pulmonary innate immune and remodeling responses in mice. J Clin Invest. 2008, 118: 2771-2784.PubMedPubMedCentral
18.
Zurück zum Zitat Moghaddam SJ, Clement CG, De la Garza MM, Zou X, Travis EL, Young HW, Evans CM, Tuvim MJ, Dickey BF: Haemophilus influenzae lysate induces aspects of the chronic obstructive pulmonary disease phenotype. Am J Respir Cell Mol Biol. 2008, 38: 629-638. 10.1165/rcmb.2007-0366OC.PubMedPubMedCentralCrossRef Moghaddam SJ, Clement CG, De la Garza MM, Zou X, Travis EL, Young HW, Evans CM, Tuvim MJ, Dickey BF: Haemophilus influenzae lysate induces aspects of the chronic obstructive pulmonary disease phenotype. Am J Respir Cell Mol Biol. 2008, 38: 629-638. 10.1165/rcmb.2007-0366OC.PubMedPubMedCentralCrossRef
19.
Zurück zum Zitat Szponar B, Pehrson C, Larsson L: Bacterial and fungal markers in tobacco smoke. Sci Total Environ. 2012, 438: 447-451.PubMedCrossRef Szponar B, Pehrson C, Larsson L: Bacterial and fungal markers in tobacco smoke. Sci Total Environ. 2012, 438: 447-451.PubMedCrossRef
20.
Zurück zum Zitat Larsson L, Pehrson C, Dechen T, Crane-Godreau M: Microbiological components in mainstream and sidestream cigarette smoke. Tob Induc Dis. 2012, 10: 13-10.1186/1617-9625-10-13.PubMedPubMedCentralCrossRef Larsson L, Pehrson C, Dechen T, Crane-Godreau M: Microbiological components in mainstream and sidestream cigarette smoke. Tob Induc Dis. 2012, 10: 13-10.1186/1617-9625-10-13.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Vernooy JH, Dentener MA, van Suylen RJ, Buurman WA, Wouters EF: Long-term intratracheal lipopolysaccharide exposure in mice results in chronic lung inflammation and persistent pathology. Am J Respir Cell Mol Biol. 2002, 26: 152-159. 10.1165/ajrcmb.26.1.4652.PubMedCrossRef Vernooy JH, Dentener MA, van Suylen RJ, Buurman WA, Wouters EF: Long-term intratracheal lipopolysaccharide exposure in mice results in chronic lung inflammation and persistent pathology. Am J Respir Cell Mol Biol. 2002, 26: 152-159. 10.1165/ajrcmb.26.1.4652.PubMedCrossRef
22.
Zurück zum Zitat Sajjan U, Ganesan S, Comstock AT, Shim J, Wang Q, Nagarkar DR, Zhao Y, Goldsmith AM, Sonstein J, Linn MJ, et al: Elastase- and LPS-exposed mice display altered responses to rhinovirus infection. Am J Physiol Lung Cell Mol Physiol. 2009, 297: L931-L944. 10.1152/ajplung.00150.2009.PubMedPubMedCentralCrossRef Sajjan U, Ganesan S, Comstock AT, Shim J, Wang Q, Nagarkar DR, Zhao Y, Goldsmith AM, Sonstein J, Linn MJ, et al: Elastase- and LPS-exposed mice display altered responses to rhinovirus infection. Am J Physiol Lung Cell Mol Physiol. 2009, 297: L931-L944. 10.1152/ajplung.00150.2009.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Sethi S, Sethi R, Eschberger K, Lobbins P, Cai X, Grant BJ, Murphy TF: Airway bacterial concentrations and exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007, 176: 356-361. 10.1164/rccm.200703-417OC.PubMedCrossRef Sethi S, Sethi R, Eschberger K, Lobbins P, Cai X, Grant BJ, Murphy TF: Airway bacterial concentrations and exacerbations of chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2007, 176: 356-361. 10.1164/rccm.200703-417OC.PubMedCrossRef
24.
Zurück zum Zitat Wilkinson TM, Hurst JR, Perera WR, Wilks M, Donaldson GC, Wedzicha JA: Effect of interactions between lower airway bacterial and rhinoviral infection in exacerbations of COPD. Chest. 2006, 129: 317-324. 10.1378/chest.129.2.317.PubMedCrossRef Wilkinson TM, Hurst JR, Perera WR, Wilks M, Donaldson GC, Wedzicha JA: Effect of interactions between lower airway bacterial and rhinoviral infection in exacerbations of COPD. Chest. 2006, 129: 317-324. 10.1378/chest.129.2.317.PubMedCrossRef
25.
Zurück zum Zitat Rohde G, Wiethege A, Borg I, Kauth M, Bauer TT, Gillissen A, Bufe A, Schultze-Werninghaus G: Respiratory viruses in exacerbations of chronic obstructive pulmonary disease requiring hospitalisation: a case–control study. Thorax. 2003, 58: 37-42. 10.1136/thorax.58.1.37.PubMedPubMedCentralCrossRef Rohde G, Wiethege A, Borg I, Kauth M, Bauer TT, Gillissen A, Bufe A, Schultze-Werninghaus G: Respiratory viruses in exacerbations of chronic obstructive pulmonary disease requiring hospitalisation: a case–control study. Thorax. 2003, 58: 37-42. 10.1136/thorax.58.1.37.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Ganesan S, Unger BL, Comstock AT, Angel KA, Mancuso P, Martinez FJ, Sajjan US: Aberrantly activated EGFR contributes to enhanced IL-8 expression in COPD airways epithelial cells via regulation of nuclear FoxO3A. Thorax. 2013, 68: 131-141. 10.1136/thoraxjnl-2012-201719.PubMedCrossRef Ganesan S, Unger BL, Comstock AT, Angel KA, Mancuso P, Martinez FJ, Sajjan US: Aberrantly activated EGFR contributes to enhanced IL-8 expression in COPD airways epithelial cells via regulation of nuclear FoxO3A. Thorax. 2013, 68: 131-141. 10.1136/thoraxjnl-2012-201719.PubMedCrossRef
27.
Zurück zum Zitat Unger BL, Faris AN, Ganesan S, Comstock AT, Hershenson MB, Sajjan US: Rhinovirus attenuates Non-typeable Hemophilus influenzae-stimulated IL-8 responses via TLR2-dependent degradation of IRAK-1. PLoS Pathog. 2012, 8: e1002969-10.1371/journal.ppat.1002969.PubMedPubMedCentralCrossRef Unger BL, Faris AN, Ganesan S, Comstock AT, Hershenson MB, Sajjan US: Rhinovirus attenuates Non-typeable Hemophilus influenzae-stimulated IL-8 responses via TLR2-dependent degradation of IRAK-1. PLoS Pathog. 2012, 8: e1002969-10.1371/journal.ppat.1002969.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Ganesan S, Faris AN, Comstock AT, Sonstein J, Curtis JL, Sajjan US: Elastase/LPS-Exposed Mice Exhibit Impaired Innate Immune Responses to Bacterial Challenge Role of Scavenger Receptor A. Am J Pathol. 2012, 180: 61-72. 10.1016/j.ajpath.2011.09.029.PubMedPubMedCentralCrossRef Ganesan S, Faris AN, Comstock AT, Sonstein J, Curtis JL, Sajjan US: Elastase/LPS-Exposed Mice Exhibit Impaired Innate Immune Responses to Bacterial Challenge Role of Scavenger Receptor A. Am J Pathol. 2012, 180: 61-72. 10.1016/j.ajpath.2011.09.029.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Chattoraj SS, Ganesan S, Jones AM, Helm JM, Comstock AT, Bright-Thomas R, LiPuma JJ, Hershenson MB, Sajjan US: Rhinovirus infection liberates planktonic bacteria from biofilm and increases chemokine responses in cystic fibrosis airway epithelial cells. Thorax. 2011, 66: 333-339. 10.1136/thx.2010.151431.PubMedPubMedCentralCrossRef Chattoraj SS, Ganesan S, Jones AM, Helm JM, Comstock AT, Bright-Thomas R, LiPuma JJ, Hershenson MB, Sajjan US: Rhinovirus infection liberates planktonic bacteria from biofilm and increases chemokine responses in cystic fibrosis airway epithelial cells. Thorax. 2011, 66: 333-339. 10.1136/thx.2010.151431.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Wang Q, Miller DJ, Bowman ER, Nagarkar DR, Schneider D, Zhao Y, Linn MJ, Goldsmith AM, Bentley JK, Sajjan US, Hershenson MB: MDA5 and TLR3 Initiate Pro-Inflammatory Signaling Pathways Leading to Rhinovirus-Induced Airways Inflammation and Hyperresponsiveness. PLoS Pathog. 2011, 7: e1002070-10.1371/journal.ppat.1002070.PubMedPubMedCentralCrossRef Wang Q, Miller DJ, Bowman ER, Nagarkar DR, Schneider D, Zhao Y, Linn MJ, Goldsmith AM, Bentley JK, Sajjan US, Hershenson MB: MDA5 and TLR3 Initiate Pro-Inflammatory Signaling Pathways Leading to Rhinovirus-Induced Airways Inflammation and Hyperresponsiveness. PLoS Pathog. 2011, 7: e1002070-10.1371/journal.ppat.1002070.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Ray P, Tang W, Wang P, Homer R, Kuhn C, Flavell RA, Elias JA: Regulated overexpression of interleukin 11 in the lung. Use to dissociate development-dependent and -independent phenotypes. J Clin Invest. 1997, 100: 2501-2511. 10.1172/JCI119792.PubMedPubMedCentralCrossRef Ray P, Tang W, Wang P, Homer R, Kuhn C, Flavell RA, Elias JA: Regulated overexpression of interleukin 11 in the lung. Use to dissociate development-dependent and -independent phenotypes. J Clin Invest. 1997, 100: 2501-2511. 10.1172/JCI119792.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Leverkoehne I, Gruber AD: The murine mCLCA3 (alias gob-5) protein is located in the mucin granule membranes of intestinal, respiratory, and uterine goblet cells. J Histochem Cytochem. 2002, 50: 829-838. 10.1177/002215540205000609.PubMedCrossRef Leverkoehne I, Gruber AD: The murine mCLCA3 (alias gob-5) protein is located in the mucin granule membranes of intestinal, respiratory, and uterine goblet cells. J Histochem Cytochem. 2002, 50: 829-838. 10.1177/002215540205000609.PubMedCrossRef
33.
Zurück zum Zitat Wedzicha JA, Seemungal TA: COPD exacerbations: defining their cause and prevention. Lancet. 2007, 370: 786-796. 10.1016/S0140-6736(07)61382-8.PubMedCrossRef Wedzicha JA, Seemungal TA: COPD exacerbations: defining their cause and prevention. Lancet. 2007, 370: 786-796. 10.1016/S0140-6736(07)61382-8.PubMedCrossRef
34.
Zurück zum Zitat Newcomb DC, Sajjan US, Nagarkar DR, Wang Q, Nanua S, Zhou Y, McHenry CL, Hennrick KT, Tsai WC, Bentley JK, et al: Human rhinovirus 1B exposure induces phosphatidylinositol 3-kinase-dependent airway inflammation in mice. Am J Respir Crit Care Med. 2008, 177: 1111-1121. 10.1164/rccm.200708-1243OC.PubMedPubMedCentralCrossRef Newcomb DC, Sajjan US, Nagarkar DR, Wang Q, Nanua S, Zhou Y, McHenry CL, Hennrick KT, Tsai WC, Bentley JK, et al: Human rhinovirus 1B exposure induces phosphatidylinositol 3-kinase-dependent airway inflammation in mice. Am J Respir Crit Care Med. 2008, 177: 1111-1121. 10.1164/rccm.200708-1243OC.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Mallia P, Message SD, Gielen V, Contoli M, Gray K, Kebadze T, Aniscenko J, Laza-Stanca V, Edwards MR, Slater L, et al: Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med. 2011, 183: 734-742. 10.1164/rccm.201006-0833OC.PubMedPubMedCentralCrossRef Mallia P, Message SD, Gielen V, Contoli M, Gray K, Kebadze T, Aniscenko J, Laza-Stanca V, Edwards MR, Slater L, et al: Experimental rhinovirus infection as a human model of chronic obstructive pulmonary disease exacerbation. Am J Respir Crit Care Med. 2011, 183: 734-742. 10.1164/rccm.201006-0833OC.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Sethi S, Maloney J, Grove L, Wrona C, Berenson CS: Airway inflammation and bronchial bacterial colonization in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006, 173: 991-998. 10.1164/rccm.200509-1525OC.PubMedPubMedCentralCrossRef Sethi S, Maloney J, Grove L, Wrona C, Berenson CS: Airway inflammation and bronchial bacterial colonization in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2006, 173: 991-998. 10.1164/rccm.200509-1525OC.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Chen R, Lim JH, Jono H, Gu XX, Kim YS, Basbaum CB, Murphy TF, Li JD: Nontypeable Haemophilus influenzae lipoprotein P6 induces MUC5AC mucin transcription via TLR2-TAK1-dependent p38 MAPK-AP1 and IKKbeta-IkappaBalpha-NF-kappaB signaling pathways. Biochem Biophys Res Commun. 2004, 324: 1087-1094. 10.1016/j.bbrc.2004.09.157.PubMedCrossRef Chen R, Lim JH, Jono H, Gu XX, Kim YS, Basbaum CB, Murphy TF, Li JD: Nontypeable Haemophilus influenzae lipoprotein P6 induces MUC5AC mucin transcription via TLR2-TAK1-dependent p38 MAPK-AP1 and IKKbeta-IkappaBalpha-NF-kappaB signaling pathways. Biochem Biophys Res Commun. 2004, 324: 1087-1094. 10.1016/j.bbrc.2004.09.157.PubMedCrossRef
38.
Zurück zum Zitat Rock JR, Randell SH, Hogan BL: Airway basal stem cells: a perspective on their roles in epithelial homeostasis and remodeling. Dis Model Mech. 2010, 3: 545-556. 10.1242/dmm.006031.PubMedPubMedCentralCrossRef Rock JR, Randell SH, Hogan BL: Airway basal stem cells: a perspective on their roles in epithelial homeostasis and remodeling. Dis Model Mech. 2010, 3: 545-556. 10.1242/dmm.006031.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Hodge S, Hodge G, Ahern J, Jersmann H, Holmes M, Reynolds PN: Smoking alters alveolar macrophage recognition and phagocytic ability: implications in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2007, 37: 748-755. 10.1165/rcmb.2007-0025OC.PubMedCrossRef Hodge S, Hodge G, Ahern J, Jersmann H, Holmes M, Reynolds PN: Smoking alters alveolar macrophage recognition and phagocytic ability: implications in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol. 2007, 37: 748-755. 10.1165/rcmb.2007-0025OC.PubMedCrossRef
40.
Zurück zum Zitat Hodge S, Matthews G, Mukaro V, Ahern J, Shivam A, Hodge G, Holmes M, Jersmann H, Reynolds PN: Cigarette smoke-induced changes to alveolar macrophage phenotype and function are improved by treatment with procysteine. Am J Respir Cell Mol Biol. 2011, 44: 673-681. 10.1165/rcmb.2009-0459OC.PubMedCrossRef Hodge S, Matthews G, Mukaro V, Ahern J, Shivam A, Hodge G, Holmes M, Jersmann H, Reynolds PN: Cigarette smoke-induced changes to alveolar macrophage phenotype and function are improved by treatment with procysteine. Am J Respir Cell Mol Biol. 2011, 44: 673-681. 10.1165/rcmb.2009-0459OC.PubMedCrossRef
41.
Zurück zum Zitat Benoit M, Desnues B, Mege JL: Macrophage polarization in bacterial infections. J Immunol. 2008, 181: 3733-3739.PubMedCrossRef Benoit M, Desnues B, Mege JL: Macrophage polarization in bacterial infections. J Immunol. 2008, 181: 3733-3739.PubMedCrossRef
42.
Zurück zum Zitat Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K: Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol. 2005, 32: 311-318. 10.1165/rcmb.2004-0309OC.PubMedCrossRef Lappalainen U, Whitsett JA, Wert SE, Tichelaar JW, Bry K: Interleukin-1beta causes pulmonary inflammation, emphysema, and airway remodeling in the adult murine lung. Am J Respir Cell Mol Biol. 2005, 32: 311-318. 10.1165/rcmb.2004-0309OC.PubMedCrossRef
43.
Zurück zum Zitat Mallia P, Footitt J, Sotero R, Jepson A, Contoli M, Trujillo-Torralbo MB, Kebadze T, Aniscenko J, Oleszkiewicz G, Gray K, et al: Rhinovirus Infection Induces Degradation of Antimicrobial Peptides and Secondary Bacterial Infection in COPD. Am J Respir Crit Care Med. 2012, 186: 1117-1124. 10.1164/rccm.201205-0806OC.PubMedPubMedCentralCrossRef Mallia P, Footitt J, Sotero R, Jepson A, Contoli M, Trujillo-Torralbo MB, Kebadze T, Aniscenko J, Oleszkiewicz G, Gray K, et al: Rhinovirus Infection Induces Degradation of Antimicrobial Peptides and Secondary Bacterial Infection in COPD. Am J Respir Crit Care Med. 2012, 186: 1117-1124. 10.1164/rccm.201205-0806OC.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Schneider D, Ganesan S, Comstock AT, Meldrum CA, Mahidhara R, Goldsmith AM, Curtis JL, Martinez FJ, Hershenson MB, Sajjan U: Increased cytokine response of rhinovirus-infected airway epithelial cells in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010, 182: 332-340. 10.1164/rccm.200911-1673OC.PubMedPubMedCentralCrossRef Schneider D, Ganesan S, Comstock AT, Meldrum CA, Mahidhara R, Goldsmith AM, Curtis JL, Martinez FJ, Hershenson MB, Sajjan U: Increased cytokine response of rhinovirus-infected airway epithelial cells in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010, 182: 332-340. 10.1164/rccm.200911-1673OC.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Lachowicz-Scroggins ME, Boushey HA, Finkbeiner WE, Widdicombe JH: Interleukin-13-induced mucous metaplasia increases susceptibility of human airway epithelium to rhinovirus infection. Am J Respir Cell Mol Biol. 2010, 43: 652-661. 10.1165/rcmb.2009-0244OC.PubMedPubMedCentralCrossRef Lachowicz-Scroggins ME, Boushey HA, Finkbeiner WE, Widdicombe JH: Interleukin-13-induced mucous metaplasia increases susceptibility of human airway epithelium to rhinovirus infection. Am J Respir Cell Mol Biol. 2010, 43: 652-661. 10.1165/rcmb.2009-0244OC.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Eddleston J, Lee RU, Doerner AM, Herschbach J, Zuraw BL: Cigarette smoke decreases innate responses of epithelial cells to rhinovirus infection. Am J Respir Cell Mol Biol. 2011, 44: 118-126. 10.1165/rcmb.2009-0266OC.PubMedPubMedCentralCrossRef Eddleston J, Lee RU, Doerner AM, Herschbach J, Zuraw BL: Cigarette smoke decreases innate responses of epithelial cells to rhinovirus infection. Am J Respir Cell Mol Biol. 2011, 44: 118-126. 10.1165/rcmb.2009-0266OC.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Hudy MH, Traves SL, Wiehler S, Proud D: Cigarette smoke modulates rhinovirus-induced airway epithelial cell chemokine production. Eur Respir J. 2010, 35: 1256-1263. 10.1183/09031936.00128809.PubMedCrossRef Hudy MH, Traves SL, Wiehler S, Proud D: Cigarette smoke modulates rhinovirus-induced airway epithelial cell chemokine production. Eur Respir J. 2010, 35: 1256-1263. 10.1183/09031936.00128809.PubMedCrossRef
48.
Zurück zum Zitat Quint JK, Donaldson GC, Goldring JJ, Baghai-Ravary R, Hurst JR, Wedzicha JA: Serum IP-10 as a biomarker of human rhinovirus infection at exacerbation of COPD. Chest. 2010, 137: 812-822. 10.1378/chest.09-1541.PubMedPubMedCentralCrossRef Quint JK, Donaldson GC, Goldring JJ, Baghai-Ravary R, Hurst JR, Wedzicha JA: Serum IP-10 as a biomarker of human rhinovirus infection at exacerbation of COPD. Chest. 2010, 137: 812-822. 10.1378/chest.09-1541.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Warwick G, Thomas PS, Yates DH: Non-invasive biomarkers in exacerbations of obstructive lung disease. Respirology. 2013, 18: 874-884. 10.1111/resp.12089.PubMedCrossRef Warwick G, Thomas PS, Yates DH: Non-invasive biomarkers in exacerbations of obstructive lung disease. Respirology. 2013, 18: 874-884. 10.1111/resp.12089.PubMedCrossRef
Metadaten
Titel
Combined exposure to cigarette smoke and nontypeable Haemophilus influenzae drives development of a COPD phenotype in mice
verfasst von
Shyamala Ganesan
Adam T Comstock
Brenton Kinker
Peter Mancuso
James M Beck
Uma S Sajjan
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Respiratory Research / Ausgabe 1/2014
Elektronische ISSN: 1465-993X
DOI
https://doi.org/10.1186/1465-9921-15-11

Weitere Artikel der Ausgabe 1/2014

Respiratory Research 1/2014 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.