Review

Animal models of chronic obstructive pulmonary disease Michael Fricker, Andrew Deane & Philip M Hansbro† 1.

Why are animal models of chronic obstructive pulmonary disease needed?

2.

COPD: what (and how) should

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

we be modelling? 3.

CS-induced models of COPD

4.

What have we learned from animal models of COPD?

5.

Modelling COPD and bacterial/ viral exacerbations of COPD

6.

Modelling systemic co-morbidities of COPD

7.

Conclusion

8.

Expert opinion



University of Newcastle and Hunter Medical Research Institute, Priority Research Centre for Asthma and Respiratory Disease, New South Wales, Australia

Introduction: Chronic obstructive pulmonary disease (COPD) is a leading global cause of mortality and chronic morbidity. Inhalation of cigarette smoke is the principal risk factor for development of this disease. COPD is a progressive disease that is typically characterised by chronic pulmonary inflammation, mucus hypersecretion, airway remodelling and emphysema that collectively reduce lung function. There are currently no therapies that effectively halt or reverse disease progression. It is hoped that the development of animal models that develop the hallmark features of COPD, in a short time frame, will aid in the identifying and testing of new therapeutic approaches. Areas covered: The authors review the recent developments in mouse models of chronic cigarette smoke-induced COPD as well as the principal findings. Furthermore, the authors discuss the use of mouse models to understand the pathogenesis and the contribution of infectious exacerbations. They also discuss the investigations of the systemic co-morbidities of COPD (pulmonary hypertension, cachexia and osteoporosis). Expert opinion: Recent advances in the field mark a point where animal models recapitulate the pathologies of COPD patients in a short time frame. They also reveal novel insights into the pathogenesis and potential treatment of this debilitating disease. Keywords: animal model, chemokine, cigarette, chronic obstructive pulmonary disease, cytokine, emphysema, guinea pig, immune system, inflammation, lung, lymphocyte, macrophage, mast cell, model, mouse, neutrophil, oxidative stress, protease, pulmonary, rat, reactive oxygen species, rodent, smoke, therapeutic, tobacco Expert Opin. Drug Discov. (2014) 9(6):629-645

Why are animal models of chronic obstructive pulmonary disease needed?

1.

Chronic obstructive pulmonary disease (COPD) is an umbrella term describing a debilitating multi-systemic disease that develops in and emanates from the lungs. It is characterised by chronic bronchitis (pulmonary inflammation), cough, airway sensory nerve sensitivity, mucus hypersecretion, airway remodelling and/or emphysema, which result in reduced lung function [1]. It is the third leading cause of death globally and is predicted to become the fifth leading cause of chronic morbidity worldwide by 2030 [1-3]. Recent studies estimate that at least 80% of COPD cases are induced by the inhalation of tobacco smoke, especially from cigarettes, with other risk factors including exposure to second-hand tobacco smoke and the fumes from burning biomass fuels, as well as genetic factors [1,4]. Despite the weight of epidemiological evidence incriminating tobacco smoke as the predominant causative factor of COPD, its incidence is predicted to rise due to continued exposure over prolonged periods coinciding with increasing uptake in populous countries (e.g., India and China) and increasing average lifespans [1]. Once initiated, the patients’ clinical condition and features of disease often deteriorate further, even after the cessation of smoking. Currently, there are no treatments that effectively 10.1517/17460441.2014.909805 © 2014 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X All rights reserved: reproduction in whole or in part not permitted

629

M. Fricker et al.

Article highlights. .

.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

.

.

Numerous independent research groups have developed cigarette smoke (CS)-exposure models in the guinea pig, rat and mouse in which animals develop the key pathological features of chronic obstructive pulmonary disease (COPD). The importance of the immune system is clear from the published findings using animal models of COPD, and several recent studies have identified novel mechanisms and drivers of the long-term disease. Progress has also been made in modelling and understanding the acute viral and bacterial exacerbations that contribute to COPD. Established CS-exposure models are now being put to good use in investigating the systemic co-morbidities associated with COPD.

This box summarises key points contained in the article.

halt the induction or progression of COPD. The use of inhaled glucocorticoids and bronchodilators to treat symptoms and exacerbations of COPD has limited clinical efficacy. Increasing our understanding of the molecular pathways and responses that contribute to the initiation and progression of COPD will facilitate the development of much-needed effective therapeutics [5]. The development of animal models of COPD that accurately recapitulate the complex multi-faceted features of this disease, particularly in a short time frame, will significantly enhance these ongoing efforts. In this review, we summarise recent advances in the development and use of animal models of experimental COPD as well as their potential limitations. This rapidly expanding field holds considerable promise, and we highlight a selection of areas of research that we consider of particular importance for future studies.

COPD: what (and how) should we be modelling? 2.

COPD is a complex condition principally defined by impaired lung function that is characterised by poorly reversible airflow limitation [1]. It is associated with a heterogeneous collection of pulmonary and extrapulmonary manifestations across patients diagnosed with various severities of disease [4]. Airflow limitation is caused by a number of pathological changes in the lungs, including chronic airway inflammation, mucus hypersecretion, airway remodelling and emphysema characterised by parenchymal destruction and loss of alveolar attachments. It is persistent chronic pulmonary and systemic inflammation that is thought to drive disease initiation and progression [5]. The decline in the condition of patients is exaggerated by exacerbations of the disease where acute increases in disease symptoms occur that are typically induced by respiratory infection with bacteria (e.g., Streptococcus 630

pneumoniae) and viruses (e.g., rhinovirus and influenza). In addition, COPD patients are more susceptible to these infections [6,7]. The chronic low-grade systemic inflammation is thought to underpin the systemic co-morbidities associated with COPD, which significantly contribute to increased morbidity rates and decreased quality of life of patients. They also have deleterious consequences for distal sites in the circulatory system, gut and skeletal muscle [8,9]. However, the mechanisms of pathogenesis in COPD are poorly understood and a further understanding of the processes involved may lead to the identification of therapeutic targets and the development of new therapies. Representative animal models are valuable in investigating these processes. They can be used to identify novel pathways of disease pathogenesis, increasing our understanding of the mechanisms and outcomes of acute bacterial and viral exacerbations and delineating the relationships between pulmonary and systemic manifestations in COPD. Many early efforts to understand the molecular basis of COPD focussed on developing animal models that reproduced individual elements of disease in isolation, such as emphysema or airway fibrosis and remodelling. These studies have identified candidate pathways that may play a role in development of these features of disease. However, it is necessary to accurately model all disease features to shed light on the true nature of their development and likely interplay [10]. An ideal animal model of COPD would reproduce a combination of the principal features of the human disease, namely chronic bronchitis, mucus hypersecretion, small airway remodelling, emphysema, impaired lung function and systemic co-morbidities. One feature that unifies the majority of COPD cases is the involvement of exposure to tobacco smoke, the major risk factor for COPD [1]. Thus, it is logical that tobacco smoke exposure would be the pertinent stimulus for modelling the induction and progression of a COPD-like state in animal models. Cigarette smoke (CS) contains a myriad of 4 -- 6000 components with known or potential modulatory effects and no other stimulus is likely to reproduce such a complexity of signals. In other diseases such as asthma, the causes are not known and must be induced by various stimuli to promote the development of features of different endotypes of the disease [11-13]. In contrast, the causes of COPD are known, and it is logical that the responses of healthy mammalian respiratory tracts of rodents and humans to CS would be similar. There is no single strong genetic predisposing factor identified and so models in wild-type animals would be ideal and development in a short time frame would be experimentally preferable. Thus, we focus the rest of our discussion on CS-induced animal models of COPD, in particular chronic CS-exposure models in which the major hallmark features of COPD, that is, airway remodelling, emphysema and impaired lung function, are induced. For discussion of alternative means of inducing COPD-like lung pathologies, readers are directed to the review by Wright et al. [10].

Expert Opin. Drug Discov. (2014) 9(6)

Animal models of COPD

Table 1. Features of COPD documented using chronic CS exposure. Species

Inflammation

Guinea pig

[18] [19] [20] [21] [28,29] [30,31] [32]

Rat

Mucus hypersecretion

Small airway remodelling

[24,25]

[22,23] [20] [21]

[26]

Emphysema

[18] [16] [20] [21] [28,29] [30,31] [32]

[32]

Impaired lung function

Smoking cessation

[22]

[17,25]

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

[139]

Mouse

[47,88] [54] [42] [140] [38] [45] [48] [127] [46]

[47,88] [54] [42] [140] [38] [45] [48] [127] [46]

[49] [84]

[46]

[46]

[47] [42] [38] [48] [127] [46]

[46]

Each row of references represents the features reported by individual research groups. COPD: Chronic obstructive pulmonary disease; CS: Cigarette smoke.

3.

CS-induced models of COPD

A number of factors must be considered with regard to the design of CS-induced models of COPD. The method of smoke production and exposure is a critical factor. ‘Mainstream’ smoke refers to the smoke that is produced and drawn back through a cigarette by the act of inhalation or ‘puffing’. This exposure occurs in active smokers. ‘Sidestream’ smoke refers to the smoke produced passively from the burning end of a cigarette that escapes into the surrounding atmosphere before being inhaled. Exposure to sidestream and exhaled mainstream smoke occurs during second-hand or passive smoke exposure. Although both types of smoke possess the same chemical composition, there is considerable difference in the relative concentrations of components [14,15]. These differences could ostensibly lead to altered cellular responses and subsequent development of COPD-like features but it remains unclear if this is indeed the case. Leberl et al.’s extensive review suggested that the two types of smoke may, in fact, not generate vastly different disease states in animal models [14]. The two systems in common use at present are ‘whole-body’ and ‘nose-only’ exposure systems. Both systems have been used extensively to provide valuable insights into COPD pathogenesis. Nose-only systems offer the benefit of reducing the potential confounding ingestion of nicotine, tar and other chemical agents by animals from their fur and are able to generate intermittent smoke exposures mimicking the puff profile of human smokers. Finally, although both systems have been reported to result in the induction of a number of different pathological features of COPD such as airway collagen deposition, emphysema and reduced lung function, Wright et al. highlighted

that nose-only exposure systems may offer an added benefit of the generation of more pronounced disease phenotypes which may make evaluation of the success of potential therapeutic interventions clearer [10]. Species, age and gender The species, age and gender of animals used are all critical considerations. Many studies have utilised guinea pigs, rats and mice as model mammals for the induction of CS-induced experimental COPD (Table 1). A limitation of the use of rodents in modelling COPD is that there are significantly fewer airway branches in their lungs compared to humans. However, as discussed below, CS-exposure studies in rodents have demonstrated important similarities in immune responses and the development of pathology to those observed in COPD patients. Thus, animal models of experimental COPD hold considerable promise for use in the development of novel therapeutic strategies. 3.1

Guinea pig Several groups have reported the development of COPD-like pathology following long-term exposure of guinea pigs to CS, with the majority of these studies originating from the laboratory of Wright and Churg. Guinea pigs develop pulmonary inflammation and increased pulmonary arterial blood pressure and muscularisation of pulmonary vessels reminiscent of that observed in COPD patients [16-21]. This is accompanied by the development of small airway remodelling and emphysema between 2 and 6 months of CS exposure and is associated with alterations in lung function such as increased resistance [21-23]. Altered mucus production in the airway epithelium has also been reported by two separate groups 3.1.1

Expert Opin. Drug Discov. (2014) 9(6)

631

M. Fricker et al.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

but this remains relatively unexplored [24-26]. Smoking cessation does not reverse pathology in the guinea pig, with increased presence of mucus secreting cells and emphysema preserved after 4 months rest following 4 months CS exposure [17,25]. Interestingly, the administration of simvastatin in this model prevented pulmonary hypertension and emphysema, but it did not affect small airway remodelling or altered lung function. This indicates that small airway remodelling and emphysema can be driven by discrete processes within the same model [27]. Rat Relatively few studies have utilised the rat as a model for the induction of CS-induced COPD-like pathology (Table 1). Several groups have demonstrated the induction of inflammation followed by emphysema [28-32]. In some cases, emphysema was observed after only 2 months of CS exposure, which represents a significantly shorter time period than commonly required in guinea pig and mouse studies [28,32]. Furthermore Wang et al. demonstrated that exposure of spontaneously hypertensive rats resulted in accelerated onset of CS-induced COPD pathology, with inflammation, increases in mucus producing cells, emphysema and impaired lung function observed within 4 weeks of CS exposure [33]. This could provide a valuable rapid onset model of disease for testing candidate therapeutic strategies, as the authors demonstrated with the phosphodiesterase-4 inhibitor rolipram. 3.1.2

Mouse Mice have been extensively studied [34] and are desirable for modelling due to their advantages of ease of breeding and maintenance, availability of genetically modified strains and reagents with which to assess molecular pathological processes as well as their relatively low cost. Studies where mice of various ages were exposed to CS demonstrated that aged mice showed reduced antioxidant responses and increased susceptibility to inflammation [35,36]. However, these studies used acute smoke exposure in which none of the chronic COPD pathologies were observed, with the exception of exaggerated inflammation. A recent study built on this early work by comparing the development of COPD pathologies in 3and 12-month-old mice exposed to CS over 6 months in a well-established model [37]. These studies confirmed a modest reduction in antioxidant responses in aged mice and demonstrated some differences in the inflammatory gene profile between young versus aged mice following smoke exposure. However, the development of COPD pathologies such as small airway remodelling and emphysema were unaffected by the age of the mice during smoke exposure [37]. Gender may also play a role in the development of the disease, as female mice appeared to develop COPD pathologies at a faster rate than male mice when exposed to identical doses of CS [38]. The differing susceptibilities of various mouse strains to CSinduced pathology have been well documented. Cavarra et al. 3.1.3

632

demonstrated that imprinting control region mice are resistant to CS-induced emphysema, and further studies supported a protective role for enhanced anti-oxidant responses in this strain [39,40]. They also showed that four susceptible strains, C57BL/6, DBA/2, B6C3F and A/J, of mice developed emphysema to different degrees following chronic CS exposure, with A/J mice being particularly susceptible [39,41-43]. These differing disease phenotypes could be viewed as an advantage as they may provide genetic clues as to why only a small portion (15%) of smokers develop COPD, as well as offer an opportunity to test the robustness of experimental findings in different strains [44]. Several research groups have now reported and characterised mouse models of CS-induced COPD (Table 1). The time required to develop COPD-like pathologies, including emphysema, varies, although the majority of studies require 6 months, and this is likely to be dependent on dose and delivery method [14,45]. Studies examining the acute inflammatory responses to short time frame (1 -- 4 week) CS exposures may provide clues as to the pathways involved in the nascent stages of COPD. However, as stand-alone studies, they do not elucidate the pathways involved in driving the long-term progression and maintenance of the disease state. Here, we focus on chronic CS-exposure models in which extensive COPD pathology is reported (e.g., emphysema, airway remodelling) and readers are directed to other recent reviews for discussion of acute CS-exposure studies [10,14,34]. We recently developed a novel mouse model of CS-induced experimental COPD. Using a nose-only system, acute and chronic onset of pulmonary inflammation was induced which lead to mucus hypersecretion and the development of airway remodelling, emphysema and impaired lung function along with systemic involvement (cachexia, loss of skeletal muscle mass, cardiac hypertrophy) [46]. We have since demonstrated airway fibrosis. This study represents a comprehensive characterisation of the induction of COPD-like pathology in mice. Importantly, emphysema and other pathological alterations were observed after only 8 weeks of CS exposure, which renders the investigation of potential mechanisms of disease and therapeutic interventions more feasible [46]. As in humans, disease persisted after the cessation of smoking (for 4 weeks). Other studies have demonstrated prolonged inflammation and emphysema following smoking cessation [38,47,48], including a study which implicated altered IL-12 and IL-10 signalling as mediators of disease maintenance and progression [48]. The smoke exposure in our model is representative of that in humans. Mice were exposed to puffs of smoke interspersed with rests from 12 cigarettes twice per day (equivalent to a pack-a-day smoker). Smoke is not forced into the mice, which breathed in the mouse lung volume equivalent doses of CS that would be comparable on a volume-to-weight basis to humans. In our model, also as in humans, features did not respond to steroid therapy and mice with experimental COPD were more susceptible to infection with S. pneumoniae and influenza.

Expert Opin. Drug Discov. (2014) 9(6)

Animal models of COPD

Table 2. Immune cell involvement in experimental COPD. Cell type

Macrophages

Neutrophils

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

Mast cell NK cell APCs gd T-cell CD3+ T-cell (CD4+ and CD8+) CD4+ T-cell CD8+ T-cell TH17 T-cell Regulatory T cell

B cell

Role

Method used

Emphysema Inflammation and emphysema Impaired efferocytosis Elastin and collagen breakdown None Inflammation, emphysema and altered lung function NK hyper-responsive phenotype, inflammation, airway damage Emphysema (IL-17 dependent) Suppression of inflammation and emphysema Pathology driving None Inflammation and emphysema Emphysema Elevated in CS-exposed mouse lung. May reduce CS-induced B-cell infiltrates Suppression of B cells reduces lymphoid follicle development and partially reduces lung tissue destruction

Preventative or therapeutic inhibition

Anti-Mac antibody Clodronate

Preventative Preventative

Anti-neutrophil antibody

Preventative and therapeutic (acute) Preventative Preventative

Anti-neutrophil antibody mMCP-6-/NKG2D-/-

Ref.

[29] [46] [75-77] [53,54] [29,46] [46] [55,56]

APC transfer gd T-cell deficient mice

Preventative

T-cell transfer

[60] [60] [58,59]

CD4+-/CD8+-/-

Preventative Preventative

[57] [57] [60] [64,65]

Cxcl13 blocking antibody

Preventative

[141]

Preventative inhibition refers to experiments in which the cell/factor has been inhibited throughout the experimental procedure to prevent the development of disease (i.e., factor-deficient mice or pharmacological agent), whereas therapeutic inhibition refers to those experiments in which the cell/factor was inhibited only for part of the experimental procedure (see references for details). APC: Antigen-presenting cell; COPD: Chronic obstructive pulmonary disease; CS: Cigarette smoke; mMCP: Mouse mast cell protease; NK: Natural killer.

3.2

Measurement of end points

The major features of COPD pathology should be evaluated in mouse models to investigate pathogenesis and the effects of interventions on the course of disease. Airway cellular inflammation can be assessed by enumerating leukocyte populations in bronchoalveolar lavage (BAL) and by FACS-based analysis of BAL cells and cell suspensions prepared directly from lung tissue [46]. This can be accompanied by determination of cytokine and chemokine mRNA and protein levels. Tissue remodelling can be evaluated using histological techniques (e.g., assessment of Masson’s Trichrome staining) to evaluate collagen deposition around airways and in the parenchyma and by measurement of breakdown products of extracellular matrix (ECM) factors [46,49]. Emphysema is commonly quantified in rodent models in terms of the mean linear intercept or destructive index, which provides discrete methods of estimating the extent of parenchymal tissue destruction [46]. A number of pathophysiologically relevant lung function changes in rodents can be measured using forced oscillation techniques (resistance, static and dynamic compliance) and forced manoeuvre techniques (FEV100: FVC that is representative of the FEV1:FVC ratio in humans, total lung capacity, work of breathing) [46]. Other less standard end points can be determined to answer specific

questions. For example, cough responses are typically assessed in guinea pigs, although a recent study demonstrated that such responses in mice can be measured using a combination of whole body plethysmography, observation and sensitive sound measurement [50].

What have we learned from animal models of COPD?

4.

Immune cell involvement COPD is characterised by chronic low-grade pulmonary and systemic inflammation, which is increased during bacterial and viral exacerbations. Inflammation is thought to underpin the progression of disease and so immune cell types and pathways have received much attention (Table 2 and Figure 1). Macrophages are well documented to be elevated in the BAL and pulmonary tissue of COPD patients, and this is also evident in animal models of CS-induced COPD [46,51]. Numerous studies have demonstrated that macrophage-associated factors such as MMP-12 [52] play critical roles in the development of pathology, implicating macrophages as important players in COPD pathogenesis (Table 3). The selective depletion of macrophages in mice provided confirmation of their importance in COPD pathogenesis, as clodronate-treated 4.1

Expert Opin. Drug Discov. (2014) 9(6)

633

M. Fricker et al.

Model of pathogenesis

Experimental observations

Human

Rodent

Cigarette smoke Smoke components

RONS, toxins Increased cell death, DAMP release

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

RONS, proteases, cytokines Inflammationinnate & adaptive immune system

Cellular damage

B-cell, CD4+ T-cell, CD8+ T-cell, γδ T-cell, TH17 T-cell T-reg cell

DAMP release

Cytokines, proteases

Macrophages, neutrophils, APCs, NKs

Cytokines proteases, RONS

Increased cell death, DAMP release

Macrophages, neutrophils, APCs, NKs, mast cells

B-cell, CD4+ T-cell, CD8+ T-cell, γδ T-cell, TH17 T-cell T-reg cell

Cell death

Tissue remodeling

Emphysema

FEV1/FVC

Decreased lung function

10+ years

COPD

FEV100/FVC, resistance, TLC, compliance

2-6 months

Figure 1. Pathogenesis of human and experimental COPD induced in response to chronic CS exposure. APC: Antigen-presenting cell; COPD: Chronic obstructive pulmonary disease; CS: Cigarette smoke; NK: Natural killer; RONS: Reactive oxygen and nitrogen species.

animals were protected from small airway remodelling, emphysema and impaired lung function in experimental COPD [46]. Similar experiments in which antibody-based depletion of neutrophils was employed demonstrated that neutrophils may contribute to early events such as exaggerated breakdown of ECM components [53,54]. Other innate immune cell types have been implicated through the use of animal models. The pathogenic role of mast cells (MCs), which was hitherto unexplored, was revealed using mice that were genetically deficient for expression of the mouse MC protease (mMCP)-6 [46]. Finally, chronic exposure to CS results in a hyper-responsive natural killer (NK) cell phenotype, which is partially suppressed in mice deficient with the activating receptor NKG2D. These mice also show reduced inflammation and airway pathology [55,56]. Recent work has implicated T cells in the pathogenesis and progression of COPD. CD8+ (but not CD4+) T-cell-deficient mice were protected from cellular inflammation as well as 634

emphysema following chronic CS exposure [57]. Recent studies have provided further evidence for the potential role of T cells as drivers of COPD pathology. Transfer of CD3+ T cells harvested from smoke-exposed mice to non-CSexposed Rag2-deficient (-/-) recipient mice induced COPD-like pathology, including increased inflammation and emphysema [58]. A follow-up study demonstrated that the transfer of CD4+ and CD8+ T cells derived from CS-exposed mice was sufficient to induce COPD immunopathology in a manner that was dependent on antigen recognition pathways, highlighting the potential involvement of autoimmune mechanisms [59]. These studies suggest that the persistent generation of pathogenic T cells by CS exposure may contribute to the chronic nature of disease, including in those individuals where disease progression continues despite cessation of smoking. Similarly, Shan et al. demonstrated that the transfer of CD1a+ antigen-presenting cells (APCs) from CS-exposed mice to naı¨ve mice was sufficient to drive

Expert Opin. Drug Discov. (2014) 9(6)

Animal models of COPD

Table 3. Factor/pathway involvement in experimental COPD. Factor/pathway

TNFR-I and -II TNFR-II

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

IL-1b

Role

Method used

Inflammation, emphysema, small airway remodelling Inflammation, emphysema and cell death

IL-1R, IL-1a

Inflammation, emphysema and small airway remodelling No role Inflammation, emphysema

IL-18/IL-18R

Inflammation, emphysema and cell death

IL-6 IFN-g EMAP-II

None Emphysema Emphysema and cell death

CCR5 CCR6

Inflammation, emphysema and cell death Lymphocyte, DC, neutrophil recruitment, emphysema, MMP-12 induction. B-cell recruitment, lymphoid aggregate development Inflammation, emphysema, lung function Inflammation, emphysema, MMP-12 induction Chemo-attraction, inflammation, emphysema Inflammation, emphysema

CXCL13 Adiponectin IL-17 ATP, P2R NE

MMP-9/-12

Inflammation, emphysema, small airway remodelling No role Inflammation, emphysema Inflammation, emphysema

Preventative

[54,68,49]

TNFR-II-/(TNFR-I-/- not protective) IL-1b-/-

Preventative

[69]

Preventative

[49]

IL-1b blocking antibody IL-1R-/-, IL-1a-/-, IL-1a blocking antibody IL-18R-/-, IL-18R blocking antibody IL-6-/IFN-g -/EMAP-II blocking antibody

Preventative Preventative

[70] [70]

Preventative

[71]

Preventative Preventative Preventative and therapeutic Preventative Preventative

[73] [82] [81]

Preventative and therapeutic Preventative Preventative

[141]

Preventative

[74]

Preventative Preventative and therapeutic Preventative Preventative

[19] [87]

Preventative Preventative Preventative

[89] [88] [90]

Preventative and therapeutic Preventative Preventative

[97]

Preventative

[75]

CCR5-/CCR6-/CLCL13 blocking antibody Adiponectin-/IL-17 Tg, IL-17-/P2Y2R-/-, P2R blockade, ATP neutralisation Inhibitor ZD0892 Inhibitor AZD9668 NE-/Inhibitor AZ11557272

MPO

iNOS

Inflammation, emphysema, prolonged emphysema, PH

SOD1 Tg mice SOD3 Tg mice, SOD3-/-, SOD3 mimetic ICR mice, MnTBAP, extracellular SOD iNOS-/-, iNOS inhibitor L-NIL

Gpx-1 Nrf-2

Suppresses inflammation Suppresses oxidative stress, emphysema, inflammation, PH. Inflammation

Inflammation, small airway remodelling, emphysema, impaired lung function, PH Reactive oxygen and Inflammation, emphysema, impaired nitrogen species efferocytosis

ABC transporters COX-2 Statin targets mTOR pathway, Rtp801 Soluble guanylyl cyclase

Ref.

TNFR-I and -II-/-

MMP-9-/MMP-12-/Inhibitor CP-471,474 MPO Inhibitor

MMP-9 MMP-12 MMPs

Preventative or therapeutic dose

Preventative and therapeutic (post emphysema) Gpx-1-/-, Gpx-1 mimic ebselen Preventative Nrf-2 activator Preventative CDDO-imidazolide, Nrf-2-/MRP1, MDR1a/b triple Preventative knockout mice Celecoxib Preventative Simvastatin Preventative Therapeutic Rtp801 Tg mice, Rtp801-/Preventative

Inflammation, emphysema Inflammation, emphysema, PH but not small airway remodelling Suppression of mTOR, oxidative stress, inflammation, emphysema Suppresses bronchial hyper-responsiveness Activator BAY58-2667 is beneficial

[82,83] [84]

[125] [60]

[86] [91]

[93] [95]

[95]

[96] [100] [142] [31] [30] [27] [98] [134]

COPD: Chronic obstructive pulmonary disease; DC: Dendritic cell; EMAP: Endothelial monocyte-activating protein; ICR: Imprinting control region; iNOS: Inducible nitrogen oxide synthase; MPO: Myeloperoxidase; mTOR: Mammalian target of rapamycin; NE: Neutrophil elastase; P2R: Purinergic receptors; SOD: Superoxide dismutase; TNFR: TNF receptor. Expert Opin. Drug Discov. (2014) 9(6)

635

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

M. Fricker et al.

COPD-like pathology, which occurred in an IL-17-dependent manner, identifying a potential therapeutic target for the treatment of chronic disease [60]. In contrast, it appeared that increases in the numbers of gd T cells and associated IL-17 production by CS exposure was important for the suppression of pathogenic TH17 responses, highlighting autoimmune aspects of COPD [60]. The link between gd T cells, IL-17 and inflammation in COPD was also described in a recent study [61]. Activation of T cells may also, in part, be mediated by the IL-1R/IL-1a-dependent accumulation of dendritic cells (DCs) in the lungs of CS-exposed mice [62]. Regulatory T cells control inflammation and are increased in humans with COPD and CS-exposed mice [63,64]. However, they clearly do not prevent pathogenesis. This suggests that these cells are dysfunctional and could be modified or further increased for therapeutic gain [65,66]. Myeloid-derived suppressor cells are also increased and altered in COPD, although the consequences of this and potential for modulation are not known [67]. Molecular drivers of COPD Given the demonstrated involvement of numerous immune cell types, it is unsurprising that numerous immune cellrelated factors have also been implicated in the pathogenesis of COPD-like disease in rodents (Table 3). 4.2

Cytokines and chemokines Churg et al. demonstrated in a series of studies that mice deficient in the expression of both TNF receptors (TNFR)-I and - II were protected from inflammation in both acute and chronic CS-exposure models and that small airway remodelling and emphysema were prevented following chronic CS exposure of TNFR double -/- mice [49,54,68]. These studies were complemented and refined independently by D’Hulst et al., who demonstrated that deletion of TNFR-II (but not TNFR-I) alone was sufficient to prevent inflammation, emphysema and associated cell death in a chronic CS-exposure model [69]. Other strongly implicated molecules are IL-17 (discussed above) and IL-1R. IL-1R is required for inflammation, small airway remodelling and emphysema following CS exposure [49,70]. Pathogenetic signalling via IL-1R involves IL-1a, as shown by using mice deficient in this factor and blocking antibodies [70]. IL-1b may also be involved in the induction and exacerbation of COPD features [46,49,70]. Expression of the IL-1 superfamily member IL-18 is elevated in the macrophages of smokers and COPD patients, a feature also observed in CS-exposed mice. Blockade of IL-18 function in IL-18R-/- mice partially suppressed inflammation, parenchymal cell death and emphysema [71]. IL-6 is increased in COPD patients and in mice exposed to CS and is required for the development of Th17 cells [72]. However, CS-induced inflammation was independent of IL-6 in mice deficient of this factor [73]. CS exposure in mice results in increased ATP release, potentially through induction of cell death, which acts as a neutrophil and macrophage chemoattractant acting 4.2.1

636

via the purinergic receptors, including P2Y2R [74]. Genetic and pharmacological blockade of this pathway reduced inflammation and emphysema following CS exposure. CS exposure may also increase ATP release through reduction of the efferocytotic capacity of macrophages, allowing dying cells to undergo secondary necrosis and plasma membrane rupture following apoptosis [75]. Impaired phagocytic ability of macrophages has also been demonstrated in COPD patients and healthy smokers, as well as smoke-exposed mice, and may contribute to disease progression [76-78]. The GM-CSF is elevated and regulates innate immune responses and cellular influx in the lung in COPD models [79,80]. Antibody-mediated neutralisation of this cytokine inhibited macrophage and neutrophil influx into the lung and the production of TNF-a and MMP-2 and -12 [79]. Endothelial monocyte activating protein II (EMAP-II) is a cell deathpromoting ligand with increased expression in the BAL of COPD patients and smokers compared to controls. EMAPII was also increased in the lungs of CS-exposed mice, and forced expression of EMAP-II was sufficient to induce cellular injury and emphysema, whereas EMAP-II neutralising antibodies prevented emphysema and cell death following chronic CS exposure [81]. The chemokine receptors CCR5 and CCR6 are also required to drive inflammation in chronic CS-exposure models and their deletion results in protection from emphysema, but not from small airway remodelling [82-84]. Interestingly, CCR6 plays a role in promoting the pulmonary infiltration of DCs and T cells, both of which are thought to play a role in chronic disease. In contrast CCR7 appears largely dispensable for the development of COPD-like symptoms in CS-exposed mice [85]. Proteolytic factors Emphysema is thought to arise at least in part due to the development of a protease/anti-protease imbalance [8,10]. Genetic deletion and pharmacological inhibition of neutrophil elastase (NE) has generated strong evidence supporting a role for this molecule in driving COPD pathogenesis [19,86,87]. Notably, a recent study in which the orally available reversible NE inhibitor AZD9668 was administered using preventative or therapeutic dosing strategies demonstrated that this may be a viable therapeutic option [87]. A seminal study using deficient mice identified MMP-12 as a driver of COPD-like disease in rodents [88], whereas MMP-9 appears dispensable [89]. Broad spectrum (CP-471,474) and relatively MMP-9/-12-specific (AZ11557272) inhibitors reduced the development of the disease when administered in a preventative manner and further studies that examine therapeutic dosing options for such agents are warranted [90,91]. Finally, we recently demonstrated that expression of the mMCP-6 was required to drive inflammation, small airway remodelling and emphysema [46]. Numbers of MCs were increased in the lungs of CS-exposed mice, and given the numerous proteolytic enzymes harboured within their secretory granules, further investigation of the role of MC factors in COPD is an interesting prospect. 4.2.2

Expert Opin. Drug Discov. (2014) 9(6)

Animal models of COPD

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

4.2.3

Oxidative stress

CS is a potent inducer of oxidative stress through the production of reactive oxygen and nitrogen species (RONS), as are the immune cells that are recruited to the lung in response to smoke exposure. RONS have important immune effector and signalling functions, but when aberrantly produced can mediate pathogenetic cellular damage, DNA alteration and cell death [92]. Several groups have investigated the use of RONS-targeting strategies for the suppression of COPDlike pathology in animal models, with promising results in some studies (Table 3). Mice overexpressing the cytosolic superoxide dismutase (SOD)1 were protected from inflammation and emphysema with reduced evidence of oxidative stress following CS exposure compared to normal air-exposed mice [93]. Yao et al. used several approaches to test the role of extracellular SOD3 to demonstrate its importance in limiting or preventing inflammation, ECM breakdown and emphysema in CS-exposed mice [94]. The implication of superoxide in oxidative stress-mediated pathology is complemented by a recent work that highlights the roles of inducible nitrogen oxide synthase (iNOS) and peroxynitrite (the production of which requires NO and superoxide) in COPD. Although several studies have shown that endothelial NOS does not play an important role in driving COPD-like pathology, iNOS knockout mice were protected from inflammation and emphysema, as well as pulmonary hypertension which developed prior to emphysema [95]. Importantly, administration of L-NIL, a selective iNOS inhibitor, prevented emphysema and pulmonary hypertension when delivered preventatively, and reversed these features when administered as a late intervention following the establishment of emphysema [95]. This finding supports the development of therapeutic agents that can suppress inflammation while stimulating regeneration of lung parenchyma. Similar to SOD, glutathione peroxidase-1 (gpx-1) is a detoxifying enzyme that also protects against ROS. Mice deficient in this factor had increased CS-induced inflammation, which could be reversed using the gpx mimetic ebselen [96]. Administration of MnTBAP, a relatively selective peroxynitrite-scavenging molecule, was able to prevent CSinduced reductions in macrophage efferocytosis function, indicating a further disease-causing mechanism associated with RONS [75]. In addition, a recent study demonstrated that administration of mannose-binding lectin was able to restore macrophage efferocytosis function in CS-exposed mice [78]. Myeloperoxidase (MPO) is a pro-oxidative enzyme involved in neutrophil and macrophage functions. MPO activity was increased following chronic CS exposure, and inhibition of MPO with both preventative and therapeutic dosing of an inhibitor (AZ1) prevented the development of several key COPD pathologies, including inflammation, small airway remodelling, emphysema and altered lung function in guinea pigs [97]. Rtp801, a suppressor of mammalian target of rapamycin (mTOR) signalling, was increased in emphysematous human lungs and in CS-exposed mice.

Overexpression of Rtp801 was sufficient to drive inflammation, oxidative stress and tissue damage in mouse lungs, and conversely Rtp801 deletion prevented CS-induced inflammation and emphysema, thus potentially uncovering an important new pathway involved in the induction of CS-induced injury [98]. In contrast, signalling by the transcription factor NF-kB, which can regulate expression of a number of immune proteins associated with COPD, was not required for inflammation in response to CS exposure [99]. Nuclear erythroid 2 p45 (Nrf2) is an oxidative stress-sensitive transcription factor that induces the expression of genes that code for antioxidants. Nrf2 pathways are reduced in COPD patients and genetic disruption of this factor in mice leads to emphysema. Treatment with an Nrf2-activating factor reduced oxidative stress, emphysema and pulmonary hypertension in CS-exposed mice, an effect which was ablated in the absence of Nrf2 expression [100]. As well as the direct effects on tissues, oxidative stress may also induce the development of auto-antibodies against host proteins such as those that are carbonyl-modified [101]. Thus, oxidative stress may also drive autoimmunity, a process that is increasingly recognised as an important component of COPD [102]. Further studies such as the recent comprehensive analysis of oxidative stress in a model of ozone-induced lung inflammation may lead to the identification of similar pathways in CS-induced disease [103]. In summary, recent studies using animal models of COPD have identified a number of exciting avenues for further exploration to develop future therapeutics.

Modelling COPD and bacterial/viral exacerbations of COPD

5.

COPD patients experience periodic exacerbations that contribute significantly to the progression of their disease. At least half of all exacerbations are caused by bacterial and viral respiratory tract infections [6,104]. Exacerbations are of particular concern, given that they correspond with increases in hospitalisations and subsequent burden on healthcare systems and are associated with increases in mortality rates [105,106]. The chronic inflammatory environment in the lungs of COPD patients is thought to alter the immune response to bacteria and viruses, resulting in acute periods of inflammation which in turn drive the progression of COPD by causing permanent tissue damage and decline in lung function. The mechanisms that cause increased susceptibility to bacterial and viral infections in COPD patients are poorly understood. Only a few studies have addressed the mechanisms associated with increased susceptibility to exacerbations in animal models of COPD (Table 4). Viral exacerbations of COPD Several groups have examined the effect of acute and chronic CS exposure on influenza virus infection. COPD patients 5.1

Expert Opin. Drug Discov. (2014) 9(6)

637

M. Fricker et al.

Table 4. Effect of CS exposure on responses to infections. Pathogen

Influenza

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

Viral mimetic poly(I:C) Staphylococcus aureus enterotoxin B Mycobacterium tuberculosis Pseudomonas aeruginosa Haemophilus influenzae Streptococcus pneumoniae Pneumocystis murina

Smoke exposure 2 weeks 6 months 4 days 5 days 8 weeks 2 weeks 14 -- 25 days 6 8 8 2

-- 8 weeks weeks weeks -- 4 months

Effects of CS exposure

Inflammation, cell death, emphysema, dependent on RNase L Inflammation Inflammation, increased viral replication Increased mortality Inflammation dependent on IL-1R and IL-1a Increased viral replication Inflammation, small airway remodelling, emphysema. Driven by IL-18, IFN-g, MAVS and RNase L Increased lymphocyte and neutrophil accumulation, IL-17, IFN-g, mucus cell hyperplasia Increased bacterial burden Increased bacterial burden and inflammation Increased bacterial burden, inflammation, weight loss Increased bacterial burden Increased susceptibility, inflammation, emphysema, impaired lung function

Ref.

[112,113] [56] [109,110] [117] [70,84] [46] [112,113] [120] [117] [118] [119] [46] [143]

CS: Cigarette smoke.

have increased susceptibility to influenza infection and experience exaggerated inflammatory immune responses following infection [107,108]. The majority of published studies report increased pulmonary and systemic inflammation following influenza infection in acute and chronic CS-exposure models. In some cases, increased viral proliferation or reduced clearance was noted [46,109]. In other cases, the dynamics of viral proliferation were not affected by CS exposure, although exaggerated inflammatory responses to influenza infection were still present [56,110]. It is possible that these discrepancies may be related to the smoke exposure protocols and the dose of virus used to induce infection. Indeed, the differential effects of CS observed were dependent on viral dose used in a chronic exposure model [111]. Elias and colleagues reported a synergistic effect of 2 weeks of CS exposure followed by influenza on inflammation and also reported increased lung damage in the form of induction of small airway remodelling, parenchymal cell death and emphysema [112,113]. The amplified immune response elicited by CS exposure followed by influenza infection may be a result of increased Toll-like receptor (TLR)3 expression observed in the lungs of mice [114]. Indeed, Kang et al. demonstrated that mice deficient in TLR3 were protected against the exaggerated inflammation and lung damage induced by influenza infection with concomitant CS exposure [112]. In addition, increased production of IL-18 was observed in CS-exposed mice following influenza infection, and genetic deletion of IL-18R was sufficient to prevent features of exacerbation such as lung inflammation and damage [112]. The same group recently demonstrated that inducible expression of IL-18 in mice was sufficient to induce inflammation and emphysema in an IFN-g-dependent manner. This supports a previous observation using poly (I:C) administration as a surrogate for viral infection in which elevated IFN-g expression was required to mediate emphysema 638

observed in a CS-exposure model of COPD with viral exacerbation [115]. In addition, a recent study revealed that the innate antiviral immune 2¢-5¢ oligoadenylate synthetase/RNaseL system is activated synergistically by CS exposure in combination with influenza infection. Deletion of RNaseL was sufficient to prevent the increases in inflammation, fibrosis, cell death and emphysema in the lungs of CS-exposed mice infected with influenza [113]. The exaggerated production of MMP-2 and -9 following influenza infection in an acute CS-exposure model may provide further clues as to the potential mechanisms of enhanced lung damage that occur in viral exacerbations of COPD [109]. Bauer et al. reported that acute (5 days) CS exposure resulted in a corticosteroid-resistant pulmonary inflammatory response to infection, although IFN-related antiviral responses were similar between groups and viral replication appeared unaffected by smoke exposure [116]. Whereas glucocorticoids were ineffective in treating the symptoms of influenza infection in smoke-exposed mice, the PPAR-g agonist pioglitazone improved resistance to viral infection in CS-exposed mice. The same research team demonstrated that blockade of IL-1a or IL-1R by genetic deletion or blocking antibody was able to prevent acute and chronic CSinduced neutrophilia in lungs, and IL-1R deletion also inhibited the amplified inflammatory response to influenza infection following acute CS exposure [70]. Thus, it is possible that strategies targeting IL-1a/IL-1R signalling or those that prevent neutrophilia could be useful in the treatment of viral exacerbations of COPD patients. Bacterial exacerbations of COPD Work investigating bacterial exacerbations in COPD using animal models is less advanced from a mechanistic perspective. However, several studies have shown increased susceptibility or inflammatory responses to infections or infection-associated 5.2

Expert Opin. Drug Discov. (2014) 9(6)

Animal models of COPD

molecules, including S. pneumoniae [46], Mycobacterium tuberculosis [117], Pseudomonas aeruginosa [118], Haemophilus influenzae [119] and Staphylococcus aureus enterotoxin B [120]. Further studies using these models are likely to uncover novel therapeutic approaches for bacterial exacerbations of COPD.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

6.

Modelling systemic co-morbidities of COPD

It is increasingly recognised that COPD manifests beyond the pulmonary system as a multi-systemic disease. COPD patients are at increased risk of developing numerous systemic co-morbidities including pulmonary hypertension (for discussion, see above and Ref. [14]), cachexia, Crohn’s disease (for discussion, see Ref. [8]) and osteoporosis [4]. Although much of the initial research with animal models of COPD has understandably focussed on the pulmonary aspects of disease, several groups are now using these established models to begin to investigate the complex interrelationship between pulmonary manifestations and systemic co-morbidities of COPD. Cachexia Cachexia is a progressive, wasting loss of adipose and skeletal muscle associated with other severe disease manifestations [121]. Approximately 25% of COPD patients develop cachexia, which is associated with around a 50% reduction in mean survival time [122]. Compromise of muscle function can occur during the early stages of disease [123]. The majority of animal studies examining COPD and cachexia have focussed on muscle, although Chen et al. reported inflammation in and depletion of white and brown adipose tissue in a chronic CS-exposure model [124]. Further indications of the potential dysregulation of adipose tissue function came from a recent study by Miller et al., who demonstrated that adiponectindeficient mice had reduced inflammation, emphysema and improved lung function compared to control mice exposed to CS [125]. Some studies report insignificant reductions in muscle mass after chronic CS exposure of mice but note a shift from type II oxidative to glycolytic muscle fibre types, reminiscent of the fibre type shift that occurs in the limb muscles of COPD patients [126,127]. Tang et al. reported loss of muscle mass and function and linked this to increased systemic release of TNF-a causing the suppression of oxygen transport to muscle and increases in atrophy-related genes MuRF1 and Atrogin-1 [128]. Caron et al. reported similar findings, although the mechanistic contribution of altered atrophy-related gene expression and signalling to muscle atrophy was not tested [129]. TNFR-II, which is required for the development of pulmonary manifestations of COPD-like disease in mice, was shown to partially contribute to the atrophy of hind limb muscles in a model of chronic CS exposure [130]. It is unclear whether this protective effect was due to reduced pulmonary or muscleTNF signalling; nevertheless this remains one of the few studies to directly test the contribution of a signalling pathway to COPD-associated cachexia.

Osteoporosis COPD patients have an increased incidence of osteoporosis, which is thought to be caused by exposure to known risk factors, including CS and systemic inflammation [131]. Akhter et al. reported reduced bone strength in mice exposed to CS over 3 months, whereas another recent study reported minor decreases in bone strength accompanied by alterations in osteoclast and osteoblast morphology and function in CS-exposed rats [132,133]. These preliminary reports suggest that animal models of CS-induced COPD could be useful in the investigation of COPD-associated osteoporosis and this merits further investigation. Many groups have developed and established models of CS-induced lung pathology. The relatively few studies examining systemic co-morbidities associated with smoking and COPD indicate that these models will likely be useful for revealing the complex mechanisms by which CS exposure and pulmonary inflammation can affect and compromise organ function throughout the body. 6.2

6.1

7.

Conclusion

Numerous studies have established chronic CS exposure as the best current method of modelling COPD pathology in rodents, although acute studies have produced valuable information and are sufficient to answer specific questions. Improvements in techniques and technologies have enabled the development of models that mimic the hallmark features of COPD pathology, including pulmonary and systemic inflammation, small airway remodelling, emphysema and impaired lung function, some within the relatively short time frames of 8 weeks. These and other models have enabled the role of several immune cells and related factors to be explored in more detail, highlighting potential new therapeutic targets. In addition, important mechanistic insights into how acute infectious exacerbations contribute to disease have also been reported. With these models firmly established, it will now be possible to investigate the molecular complexities underlying the development of systemic co-morbidities, as well as develop novel therapeutic agents for the treatment of COPD. 8.

Expert opinion

Great strides have been made in the development of animal models of CS-induced COPD. These models variously reproduce many of the pathophysiological features of COPD, including aberrant inflammation, small airway remodelling, emphysema and impaired lung function, as well as dysfunctional immune responses in response to viral or bacterial infection. However, some studies are limited in terms of the scope of aspects of disease reported. There is unlikely to be complete standardisation of protocols, equipment and animals between laboratories [14]. Nevertheless, recent models

Expert Opin. Drug Discov. (2014) 9(6)

639

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

M. Fricker et al.

have been developed that accurately reproduce the hallmark features of human disease in a reasonable time frame [46]. This is perhaps unsurprising as, unlike in other diseases, the causes of COPD are known and it is logical that exposure of the healthy murine respiratory tract to CS will elicit similar responses and lead to the development of similar disease features compared to those in humans. The field appears to be at a point now where researchers are no longer simply striving to replicate disease pathology, and are in a position to utilise these models to provide genuine insights into the pathogenesis of COPD and identify novel therapeutic strategies. The use of adoptive transfer of immune cell types from CS-exposed to non-CS-exposed mice has offered some tantalising hints about the roles of the adaptive immune system and autoimmunity in driving chronic COPD [58,59]. Furthermore, the identification of novel cell types and the factors they produce that may contribute to pathogenesis such as (MCs) and TH17 cells opens up many possibilities for further investigation [46,60]. The use of a pharmacological agents to not only halt but also reverse emphysema in mouse CS-exposure models highlights their exciting potential for drug discovery and target identification [95]. This also raises the prospect of focussing on pathways that might be activated for therapeutic gain, as has recently been shown with a small molecule activator of soluble guanylyl cyclase and in the administration of adiposederived stem cells to suppress CS-induced inflammation and Bibliography Papers of special note have been highlighted as either of interest () or of considerable interest () to readers. 1.

2.

3.

4.

5.

640

Global initiative for chronic Obstructive Lung Disease (GOLD). Global Strategy for the Diagnosis, Management and Prevention of COPD. Available from: http://www.goldcopdorg/2013 World Health Organisation. The global burden of disease: 2004 update. 2004. Available from: http://www.hoint/ healthinfo/global_burden_disease/ 2004_report_update/en/indexhtml Lozano R, Naghavi M, Foreman K, et al. Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012;15;380(9859): 2095-128

6.

7.

8.

9.

injury [134,135]. Another area in which animal models will be valuable is in the investigation of corticosteroid insensitivity that characterises inflammation associated with COPD. Several studies have shown that CS-induced inflammation (both acute and chronic) in rodents is steroid-insensitive [46,136]. Investigations of acute CS-exposure mouse models have identified phosphatidylinositol 3-kinase d (PI3Kd) to play an important role. Inhibition of PI3Kd prevents CS exposure-induced decreases in histone deacetylase 2 resulting in the restoration of steroid responsiveness of airway inflammation [136]. Recent data also implicates the elevation of serum amyloid A as a contributing factor to the generation of corticosteroid refractory inflammation in COPD [137]. Further studies are required to investigate other avenues for restoring steroid sensitivity, and the long-term efficacy of such approaches could be assessed in chronic CS-exposure models [138].

Declaration of interest The authors were supported by the National Health and Medical Research Council, Australia. The authors have no other relevant affiliations or financial involvement with any organisation or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Bauer CM, Morissette MC, Stampfli MR. The influence of cigarette smoking on viral infections: translating bench science to impact COPD pathogenesis and acute exacerbations of COPD clinically. Chest 2013;143(1):196-206 Wedzicha JA, Seemungal TA. COPD exacerbations: defining their cause and prevention. Lancet 2007;370(9589):786-96 Keely S, Talley NJ, Hansbro PM. Pulmonary-intestinal cross-talk in mucosal inflammatory disease. Mucosal Immunol 2012;5(1):7-18 Hansen MJ, Gualano RC, Bozinovski S, et al. Therapeutic prospects to treat skeletal muscle wasting in COPD (chronic obstructive lung disease). Pharmacol Ther 2006;109(1-2):162-72

Decramer M, Janssens W. Chronic obstructive pulmonary disease and comorbidities. Lancet Resp Med 2013;1(1):73-83

10.

Wright JL, Cosio M, Churg A. Animal models of chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2008;295(1):L1-15

Barnes PJ. New anti-inflammatory targets for chronic obstructive pulmonary disease. Nat Rev Drug Discov 2013;12(7):543-59

11.

Thorburn AN, Foster PS, Gibson PG, Hansbro PM. Components of Streptococcus pneumoniae suppress allergic airways disease and NKT cells by

Expert Opin. Drug Discov. (2014) 9(6)

inducing regulatory T cells. J Immunol 2012;188(9):4611-20 12.

Essilfie AT, Simpson JL, Dunkley ML, et al. Combined Haemophilus influenzae respiratory infection and allergic airways disease drives chronic infection and features of neutrophilic asthma. Thorax 2012;67(7):588-99

13.

Starkey MR, Jarnicki AG, Essilfie AT, et al. Murine models of infectious exacerbations of airway inflammation. Curr Opin Pharmacol 2013;13(3):337-44

14.

Leberl M, Kratzer A, Taraseviciene-Stewart L. Tobacco smoke induced COPD/emphysema in the animal model-are we all on the same page? Front Physiol 2013;4:91

15.

Moritsugu KP. The 2006 Report of the Surgeon General: the health consequences of involuntary exposure to tobacco smoke. Am J Prev Med 2007;32(6):542-3

16.

Wright JL, Churg A. Cigarette smoke causes physiologic and morphologic changes of emphysema in the guinea pig. Am Rev Respir Dis 1990;142(6 Pt 1):1422-8

Animal models of COPD

17.

Wright JL, Sun JP. Effect of smoking cessation on pulmonary and cardiovascular function and structure: analysis of guinea pig model. J Appl Physiol 1994;76(5):2163-8

18.

Selman M, Montano M, Ramos C, et al. Tobacco smoke-induced lung emphysema in guinea pigs is associated with increased interstitial collagenase. Am J Physiol 1996;271(5 Pt 1):L734-43

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

19.

20.

21.

22.

23.

24.

25.

26.

27.

.

Wright JL, Farmer SG, Churg A. Synthetic serine elastase inhibitor reduces cigarette smoke-induced emphysema in guinea pigs. Am J Respir Crit Care Med 2002;166(7):954-60 Golovatch P, Mercer BA, Lemaitre V, et al. Role for cathepsin K in emphysema in smoke-exposed guinea pigs. Exp Lung Res 2009;35(8):631-45 Dominguez-Fandos D, Peinado VI, Puig-Pey R, et al. Pulmonary inflammatory reaction and structural changes induced by cigarette smoke exposure in the Guinea pig. COPD 2012;9(5):473-84 Wright JL, Postma DS, Kerstjens HA, et al. Airway remodeling in the smoke exposed guinea pig model. Inhal Toxicol 2007;19(11):915-23

remodelling is unaffected by simvastin treatment, indicating discrete pathogenic mechanisms for various pulmonary pathologies associated with chronic obstructive pulmonary disease (COPD). 28.

Ofulue AF, Ko M, Abboud RT. Time course of neutrophil and macrophage elastinolytic activities in cigarette smoke-induced emphysema. Am J Physiol 1998;275(6 Pt 1): L1134-44

29.

Ofulue AF, Ko M. Effects of depletion of neutrophils or macrophages on development of cigarette smoke-induced emphysema. Am J Physiol 1999;277(1 Pt 1):L97-105

30.

Lee JH, Lee DS, Kim EK, et al. Simvastatin inhibits cigarette smokinginduced emphysema and pulmonary hypertension in rat lungs. Am J Respir Crit Care Med 2005;172(8):987-93

31.

Roh GS, Yi CO, Cho YJ, et al. Anti-inflammatory effects of celecoxib in rat lungs with smoke-induced emphysema. Am J Physiol Lung Cell Mol Physiol 2010;299(2):L184-91

38.

March TH, Wilder JA, Esparza DC, et al. Modulators of cigarette smokeinduced pulmonary emphysema in A/J mice. Toxicol Sci 2006;92(2):545-59

39.

Cavarra E, Bartalesi B, Lucattelli M, et al. Effects of cigarette smoke in mice with different levels of alpha(1)proteinase inhibitor and sensitivity to oxidants. Am J Respir Crit Care Med 2001;1;164(5):886-90

40.

Rangasamy T, Cho CY, Thimmulappa RK, et al. Genetic ablation of Nrf2 enhances susceptibility to cigarette smoke-induced emphysema in mice. J Clin Invest 2004;114(9):1248-59

41.

Bartalesi B, Cavarra E, Fineschi S, et al. Different lung responses to cigarette smoke in two strains of mice sensitive to oxidants. Eur Respir J 2005;25(1):15-22

42.

Foronjy RF, Mercer BA, Maxfield MW, et al. Structural emphysema does not correlate with lung compliance: lessons from the mouse smoking model. Exp Lung Res 2005;31(6):547-62

43.

March TH, Bowen LE, Finch GL, et al. Effects of strain and treatment with inhaled aII-trans-retinoic acid on cigarette smoke-induced pulmonary emphysema in mice. COPD 2005;2(3):289-302

32.

Wright JL, Churg A. Smoke-induced emphysema in guinea pigs is associated with morphometric evidence of collagen breakdown and repair. Am J Physiol 1995;268(1 Pt 1):L17-20

Kratzer A, Salys J, Nold-Petry C, et al. Role of IL-18 in second-hand smokeinduced emphysema. Am J Respir Cell Mol Biol 2013;48(6):725-32

33.

44.

Wright JL, Ngai T, Churg A. Effect of long-term exposure to cigarette smoke on the small airways of the guinea pig. Exp Lung Res 1992;18(1):105-14

Guerassimov A, Hoshino Y, Takubo Y, et al. The development of emphysema in cigarette smoke-exposed mice is strain dependent. Am J Respir Crit Care Med 2004;170(9):974-80

.

Wang L, Yang J, Guo L, et al. Use of a soluble epoxide hydrolase inhibitor in smoke-induced chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 2012;46(5):614-22 Use of spontaneously hypertensive rat to induce a range of COPD pathologies in a short 4-week CS-exposure model.

45.

Hodge-Bell KC, Lee KM, Renne RA, et al. Pulmonary inflammation in mice exposed to mainstream cigarette smoke. Inhal Toxicol 2007;19(4):361-76

34.

Vlahos R, Bozinovski S. Recent advances in pre-clinical mouse models of COPD. Clin Sci (Lond) 2014;126(4):253-65

46.

35.

Moriyama C, Betsuyaku T, Ito Y, et al. Aging enhances susceptibility to cigarette smoke-induced inflammation through bronchiolar chemokines. Am J Respir Cell Mol Biol 2010;42(3):304-11

Beckett EL, Stevens RL, Jarnicki AG, et al. A new short-term mouse model of chronic obstructive pulmonary disease identifies a role for mast cell tryptase in pathogenesis. J Allergy Clin Immunol 2013;131(3):752-62 Short time frame model demonstrated multiple features of COPD pathology. Involvement of mast cells was demonstrated for the first time.

Wright JL, Churg A. Smoking cessation decreases the number of metaplastic secretory cells in the small airways of the Guinea pig. Inhal Toxicol 2002;14(11):1153-9 Ardite E, Peinado VI, Rabinovich RA, et al. Systemic effects of cigarette smoke exposure in the guinea pig. Respir Med 2006;100(7):1186-94 Wright JL, Zhou S, Preobrazhenska O, et al. Statin reverses smoke-induced pulmonary hypertension and prevents emphysema but not airway remodeling. Am J Respir Crit Care Med 2011;183(1):50-8 Therapeutic dosing of simvastin prevents cigarette smoke (CS)-induced emphysema and pulmonary hypertension. Small airway

36.

37.

Gould NS, Min E, Gauthier S, et al. Aging adversely affects the cigarette smoke-induced glutathione adaptive response in the lung. Am J Respir Crit Care Med 2010;182(9):1114-22 Zhou S, Wright JL, Liu J, et al. Aging does not Enhance Experimental Cigarette Smoke-Induced COPD in the Mouse. PLoS One 2013;8(8):e71410

Expert Opin. Drug Discov. (2014) 9(6)

..

47.

Nakanishi Y, Kobayashi D, Asano Y, et al. Clarithromycin prevents smokeinduced emphysema in mice. Am J Respir Crit Care Med 2009;179(4):271-8

48.

Braber S, Henricks PA, Nijkamp FP, et al. Inflammatory changes in the airways of mice caused by cigarette

641

M. Fricker et al.

smoke exposure are only partially reversed after smoking cessation. Respir Res 2010;11:99 49.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

50.

51.

Churg A, Zhou S, Wang X, et al. The role of interleukin-1beta in murine cigarette smoke-induced emphysema and small airway remodeling. Am J Respir Cell Mol Biol 2009;40(4):482-90 Chen L, Lai K, Lomask JM, et al. Detection of mouse cough based on sound monitoring and respiratory airflow waveforms. PLoS One 2013;8(3):e59263

Brusselle GG. Matrix metalloproteinase 12, asthma, and COPD. N Engl J Med 2009;361(27):2664-5

53.

Dhami R, Gilks B, Xie C, et al. Acute cigarette smoke-induced connective tissue breakdown is mediated by neutrophils and prevented by alpha1-antitrypsin. Am J Respir Cell Mol Biol 2000;22(2):244-52

55.

56.

57.

58.

.

59.

642

60.

..

Barnes PJ. Alveolar macrophages as orchestrators of COPD. COPD 2004;1(1):59-70

52.

54.

.

Churg A, Wang RD, Tai H, et al. Tumor necrosis factor-alpha drives 70% of cigarette smoke-induced emphysema in the mouse. Am J Respir Crit Care Med 2004;170(5):492-8 Motz GT, Eppert BL, Wortham BW, et al. Chronic cigarette smoke exposure primes NK cell activation in a mouse model of chronic obstructive pulmonary disease. J Immunol 2010;184(8):4460-9 Wortham BW, Eppert BL, Motz GT, et al. NKG2D mediates NK cell hyperresponsiveness and influenzainduced pathologies in a mouse model of chronic obstructive pulmonary disease. J Immunol 2012;188(9):4468-75 Maeno T, Houghton AM, Quintero PA, et al. CD8+ T Cells are required for inflammation and destruction in cigarette smoke-induced emphysema in mice. J Immunol 2007;178(12):8090-6 Motz GT, Eppert BL, Wesselkamper SC, et al. Chronic cigarette smoke exposure generates pathogenic T cells capable of driving COPD-like disease in Rag2-/mice. Am J Respir Crit Care Med 2010;181(11):1223-33 Transfer of CD3+ T cells from CS-exposed mice can drive COPD-like pathology in non-CS-exposed recipient mice. Eppert BL, Wortham BW, Flury JL, Borchers MT. Functional characterization

of T cell populations in a mouse model of chronic obstructive pulmonary disease. J Immunol 2013;190(3):1331-40 Implication of autoimmune mechanisms in the induction of COPD-like pathology by T cells. Shan M, Yuan X, Song LZ, et al. Cigarette smoke induction of osteopontin (SPP1) mediates T(H)17 inflammation in human and experimental emphysema. Sci Transl Med 2012;4(117):117ra9 Transfer of APCs from CS-exposed mice to naı¨ve mice can drive emphysema. Identification of TH17 cells as pathology-driving while gd T cells suppress pathology.

61.

Anthony D, Seow HJ, Uddin M, et al. Serum amyloid A promotes lung neutrophilia by increasing IL-17A levels in the mucosa and gammadelta T cells. Am J Respir Crit Care Med 2013;188(2):179-86

62.

Botelho FM, Nikota JK, Bauer CM, et al. Cigarette smoke-induced accumulation of lung dendritic cells is interleukin-1alpha-dependent in mice. Respir Res 2012;13:81

63.

Dancer R, Sansom DM. Regulatory T cells and COPD. Thorax 2013;68(12):1176-8

64.

Demoor T, Bracke KR, Joos GF, Brusselle GG. Increased T-regulatory cells in lungs and draining lymph nodes in a murine model of COPD. Eur Respir J 2010;35(3):688-9

65.

Brandsma CA, Hylkema MN, van der Strate BW, et al. Heme oxygenase-1 prevents smoke induced B-cell infiltrates: a role for regulatory T cells? Respir Res 2008;9:17

66.

Thorburn AN, Hansbro PM. Harnessing regulatory T cells to suppress asthma: from potential to therapy. Am J Respir Cell Mol Biol 2010;43(5):511-19

67.

68.

69.

receptor p75 in cigarette smoke-induced pulmonary inflammation and emphysema. Eur Respir J 2006;28(1):102-12 70.

.

Botelho FM, Bauer CM, Finch D, et al. IL-1alpha/IL-1R1 expression in chronic obstructive pulmonary disease and mechanistic relevance to smoke-induced neutrophilia in mice. PLoS One 2011;6(12):e28457 Implication of IL-1 signaling in the pathogenesis of COPD and the altered immune response to influenza infection caused by CS exposure.

71.

Kang MJ, Homer RJ, Gallo A, et al. IL-18 is induced and IL-18 receptor alpha plays a critical role in the pathogenesis of cigarette smoke-induced pulmonary emphysema and inflammation. J Immunol 2007;178(3):1948-59

72.

Barnes PJ. The cytokine network in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 2009;41(6):631-8

73.

Pauwels NS, Bracke KR, Maes T, et al. The role of interleukin-6 in pulmonary and systemic manifestations in a murine model of chronic obstructive pulmonary disease. Exp Lung Res 2010;36(8):469-83

74.

Cicko S, Lucattelli M, Muller T, et al. Purinergic receptor inhibition prevents the development of smoke-induced lung injury and emphysema. J Immunol 2010;185(1):688-97 Several methods are employed by the authors to demonstrate the pathogenic role of purinergic receptors in experimental COPD.

.

75.

Scrimini S, Pons J, Agusti A, et al. Differential effects of smoking and COPD upon circulating myeloid derived suppressor cells. Respir Med 2013;107(12):1895-903

Richens TR, Linderman DJ, Horstmann SA, et al. Cigarette smoke impairs clearance of apoptotic cells through oxidant-dependent activation of RhoA. Am J Respir Crit Care Med 2009;179(11):1011-21

76.

Churg A, Dai J, Tai H, et al. Tumor necrosis factor-alpha is central to acute cigarette smoke-induced inflammation and connective tissue breakdown. Am J Respir Crit Care Med 2002;166(6):849-54

Hodge S, Hodge G, Ahern J, et al. Smoking alters alveolar macrophage recognition and phagocytic ability: implications in chronic obstructive pulmonary disease. Am J Respir Cell Mol Biol 2007;37(6):748-55

77.

Hodge S, Matthews G, Mukaro V, et al. Cigarette smoke-induced changes to alveolar macrophage phenotype and function are improved by treatment with procysteine. Am J Respir Cell Mol Biol 2011;44(5):673-81

D’Hulst AI, Bracke KR, Maes T, et al. Role of tumour necrosis factor-alpha

Expert Opin. Drug Discov. (2014) 9(6)

Animal models of COPD

78.

.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

79.

80.

81.

.

82.

83.

84.

85.

86.

87.

Hodge S, Matthews G, Dean MM, et al. Therapeutic role for mannose-binding lectin in cigarette smoke-induced lung inflammation? Evidence from a murine model. Am J Respir Cell Mol Biol 2010;42(2):235-42 Adminstration of mannose-binding lectin restores efferocytosis and reduces inflammation in CS-exposed mice, correlated with human patient data. Vlahos R, Bozinovski S, Hamilton JA, Anderson GP. Therapeutic potential of treating chronic obstructive pulmonary disease (COPD) by neutralising granulocyte macrophage-colony stimulating factor (GM-CSF). Pharmacol Ther 2006;112(1):106-15 Saha S, Doe C, Mistry V, et al. Granulocyte-macrophage colonystimulating factor expression in induced sputum and bronchial mucosa in asthma and COPD. Thorax 2009;64(8):671-6 Clauss M, Voswinckel R, Rajashekhar G, et al. Lung endothelial monocyteactivating protein 2 is a mediator of cigarette smoke-induced emphysema in mice. J Clin Invest 2011;121(6):2470-9 Authors used a range of methods to demonstrate that endothelial monocyte activating protein-II plays a causative role in experimental COPD. Ma B, Kang MJ, Lee CG, et al. Role of CCR5 in IFN-gamma-induced and cigarette smoke-induced emphysema. J Clin Invest 2005;115(12):3460-72 Bracke KR, D’Hulst A I, Maes T, et al. Cigarette smoke-induced pulmonary inflammation, but not airway remodelling, is attenuated in chemokine receptor 5-deficient mice. Clin Exp Allergy 2007;37(10):1467-79

characterization of a novel oral inhibitor of neutrophil elastase. J Pharmacol Exp Ther 2011;339(1):313-20 88.

Hautamaki RD, Kobayashi DK, Senior RM, Shapiro SD. Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice. Science 1997;277(5334):2002-4

89.

Atkinson JJ, Lutey BA, Suzuki Y, et al. The role of matrix metalloproteinase-9 in cigarette smoke-induced emphysema. Am J Respir Crit Care Med 2011;183(7):876-84

90.

Selman M, Cisneros-Lira J, Gaxiola M, et al. Matrix metalloproteinases inhibition attenuates tobacco smokeinduced emphysema in Guinea pigs. Chest 2003;123(5):1633-41

91.

Churg A, Wang R, Wang X, et al. Effect of an MMP-9/MMP-12 inhibitor on smoke-induced emphysema and airway remodelling in guinea pigs. Thorax 2007;62(8):706-13

92.

93.

Foronjy R, D’Armiento J. The effect of cigarette smoke-derived oxidants on the inflammatory response of the lung. Clin Appl Immunol Rev 2006;1;6(1):53-72

94.

Yao H, Arunachalam G, Hwang JW, et al. Extracellular superoxide dismutase protects against pulmonary emphysema by attenuating oxidative fragmentation of ECM. Proc Natl Acad Sci USA 2010;31;107(35):15571-6 A combination of genetic and pharmacological approaches are utilised to demonstrate the protective role of the antioxidant enzyme superoxide dismutase 3 in experimental COPD, correlated with human COPD patient data.

..

Bracke KR, D’Hulst A I, Maes T, et al. Cigarette smoke-induced pulmonary inflammation and emphysema are attenuated in CCR6-deficient mice. J Immunol 2006;177(7):4350-9 Demoor T, Bracke KR, Vermaelen KY, et al. CCR7 modulates pulmonary and lymph node inflammatory responses in cigarette smoke-exposed mice. J Immunol 2009;183(12):8186-94 Shapiro SD, Goldstein NM, Houghton AM, et al. Neutrophil elastase contributes to cigarette smoke-induced emphysema in mice. Am J Pathol 2003;163(6):2329-35 Stevens T, Ekholm K, Granse M, et al. AZD9668: pharmacological

Caramori G, Adcock IM, Casolari P, et al. Unbalanced oxidant-induced DNA damage and repair in COPD: a link towards lung cancer. Thorax 2011;66(6):521-7

95.

..

96.

Seimetz M, Parajuli N, Pichl A, et al. Inducible NOS inhibition reverses tobacco-smoke-induced emphysema and pulmonary hypertension in mice. Cell 2011;14;147(2):293-305 First publication to demonstrate that a pharmacological intervention can halt and reverse established emphysema in a CS-induced model of COPD. Duong C, Seow HJ, Bozinovski S, et al. Glutathione peroxidase-1 protects against cigarette smoke-induced lung Expert Opin. Drug Discov. (2014) 9(6)

inflammation in mice. Am J Physiol Lung Cell Mol Physiol 2010;299(3):L425-33 97.

.

Churg A, Marshall CV, Sin DD, et al. Late intervention with a myeloperoxidase inhibitor stops progression of experimental chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2012;185(1):34-43 Therapeutic dosing with a myeloperoxidase inhibitor prevents development of COPD-like pathology, including emphysema.

98.

Yoshida T, Mett I, Bhunia AK, et al. Rtp801, a suppressor of mTOR signaling, is an essential mediator of cigarette smoke-induced pulmonary injury and emphysema. Nat Med 2010;16(7):767-73

99.

Rastrick JM, Stevenson CS, Eltom S, et al. Cigarette smoke induced airway inflammation is independent of NF-kappaB signalling. PLoS One 2013;8(1):e54128

100. Sussan TE, Rangasamy T, Blake DJ, et al. Targeting Nrf2 with the triterpenoid CDDO-imidazolide attenuates cigarette smoke-induced emphysema and cardiac dysfunction in mice. Proc Natl Acad Sci USA 2009;6;106(1):250-5 . Activation of Nrf2 inhibits oxidative stress, pulmonary damage and inflammation and pulmonary hypertension associated with chronic CS exposure. 101. Kirkham PA, Caramori G, Casolari P, et al. Oxidative stress-induced antibodies to carbonyl-modified protein correlate with severity of chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2011;184(7):796-802 102. Kheradmand F, Shan M, Xu C, Corry DB. Autoimmunity in chronic obstructive pulmonary disease: clinical and experimental evidence. Expert Rev Clin Immunol 2012;8(3):285-92 103. Wiegman CH, Li F, Clarke CJ, et al. A comprehensive analysis of oxidative stress in the ozone-induced lung inflammation mouse model. Clin Sci (Lond) 2014;126(6):425-40 104. Wedzicha JA. Role of viruses in exacerbations of chronic obstructive pulmonary disease. Proc Am Thorac Soc 2004;1(2):115-20 105. Wedzicha JA. Airway infection accelerates decline of lung function in chronic

643

M. Fricker et al.

obstructive pulmonary disease. Am J Respir Crit Care Med 2001;164(10 Pt 1):1757-8

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

106. Wesseling G. Occasional review: influenza in COPD: pathogenesis, prevention, and treatment. Int J Chron Obstruct Pulmon Dis 2007;2(1):5-10 107. Seemungal T, Harper-Owen R, Bhowmik A, et al. Respiratory viruses, symptoms, and inflammatory markers in acute exacerbations and stable chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2001;164(9):1618-23 108. Hsu AC, See HV, Hansbro PM, Wark PA. Innate immunity to influenza in chronic airways diseases. Respirology 2012;17(8):1166-75 109. Gualano RC, Hansen MJ, Vlahos R, et al. Cigarette smoke worsens lung inflammation and impairs resolution of influenza infection in mice. Respir Res 2008;9:53 110. Yageta Y, Ishii Y, Morishima Y, et al. Role of Nrf2 in host defense against influenza virus in cigarette smokeexposed mice. J Virol 2011;85(10):4679-90 111. Robbins CS, Bauer CM, Vujicic N, et al. Cigarette smoke impacts immune inflammatory responses to influenza in mice. Am J Respir Crit Care Med 2006;174(12):1342-51 112. Kang MJ, Lee CG, Lee JY, et al. Cigarette smoke selectively enhances viral PAMP- and virus-induced pulmonary innate immune and remodeling responses in mice. J Clin Invest 2008;118(8):2771-84 . Demonstration of synergistic effect of CS exposure with influenza infection to accelerate development of COPD-like pathology. 113. Zhou Y, Kang MJ, Jha BK, et al. Role of ribonuclease L in viral pathogenassociated molecular pattern/influenza virus and cigarette smoke-induced inflammation and remodeling. J Immunol 2013;191(5):2637-46 . Identification of novel pathways involved in the exagerated immune response elicited by a combination of CS exposure and influenza infection. 114. Kimura G, Ueda K, Eto S, et al. Toll-like receptor 3 stimulation causes corticosteroid-refractory airway neutrophilia and hyperresponsiveness in mice. Chest 2013;144(1):99-105

644

115. Kang MJ, Choi JM, Kim BH, et al. IL-18 induces emphysema and airway and vascular remodeling via IFN-gamma, IL-17A, and IL-13. Am J Respir Crit Care Med 2012;185(11):1205-17 116. Bauer CM, Zavitz CC, Botelho FM, et al. Treating viral exacerbations of chronic obstructive pulmonary disease: insights from a mouse model of cigarette smoke and H1N1 influenza infection. PLoS One 2010;5(10):e13251

126. Gosker HR, Langen RC, Bracke KR, et al. Extrapulmonary manifestations of chronic obstructive pulmonary disease in a mouse model of chronic cigarette smoke exposure. Am J Respir Cell Mol Biol 2009;40(6):710-16 127. Rinaldi M, Maes K, De Vleeschauwer S, et al. Long-term nose-only cigarette smoke exposure induces emphysema and mild skeletal muscle dysfunction in mice. Dis Model Mech 2012;5(3):333-41

117. Feng Y, Kong Y, Barnes PF, et al. Exposure to cigarette smoke inhibits the pulmonary T-cell response to influenza virus and Mycobacterium tuberculosis. Infect Immun 2011;79(1):229-37

128. Tang K, Wagner PD, Breen EC. TNF-alpha-mediated reduction in PGC-1alpha may impair skeletal muscle function after cigarette smoke exposure. J Cell Physiol 2010;222(2):320-7

118. Drannik AG, Pouladi MA, Robbins CS, et al. Impact of cigarette smoke on clearance and inflammation after Pseudomonas aeruginosa infection. Am J Respir Crit Care Med 2004;170(11):1164-71

129. Caron MA, Morissette MC, Theriault ME, et al. Alterations in skeletal muscle cell homeostasis in a mouse model of cigarette smoke exposure. PLoS One 2013;8(6):e66433

120. Huvenne W, Lanckacker EA, Krysko O, et al. Exacerbation of cigarette smokeinduced pulmonary inflammation by Staphylococcus aureus enterotoxin B in mice. Respir Res 2011;12:69

130. De Paepe B, Brusselle GG, Maes T, et al. TNF alpha receptor genotype influences smoking-induced muscle-fibre-type shift and atrophy in mice. Acta Neuropathol 2008;115(6):675-81 . This paper is one of the few studies which establishes a causative relationship between a signalling pathway (TNF-a) and muscular dysfunction in COPD.

121. Sakuma K, Yamaguchi A. Sarcopenia and cachexia: the adaptations of negative regulators of skeletal muscle mass. J Cachexia Sarcopenia Muscle 2012;3(2):77-94

131. Romme EA, Smeenk FW, Rutten EP, Wouters EF. Osteoporosis in chronic obstructive pulmonary disease. Expert Rev Respir Med 2013;7(4):397-410

122. Wagner PD. Possible mechanisms underlying the development of cachexia in COPD. Eur Respir J 2008;31(3):492-501

132. Akhter MP, Lund AD, Gairola CG. Bone biomechanical property deterioration due to tobacco smoke exposure. Calcif Tissue Int 2005;77(5):319-26

119. Gaschler GJ, Skrtic M, Zavitz CC, et al. Bacteria challenge in smoke-exposed mice exacerbates inflammation and skews the inflammatory profile. Am J Respir Crit Care Med 2009;179(8):666-75

123. Seymour JM, Spruit MA, Hopkinson NS, et al. The prevalence of quadriceps weakness in COPD and the relationship with disease severity. Eur Respir J 2010;36(1):81-8 124. Chen H, Hansen MJ, Jones JE, et al. Long-term cigarette smoke exposure increases uncoupling protein expression but reduces energy intake. Brain Res 2008;1228:81-8 125. Miller M, Pham A, Cho JY, et al. Adiponectin-deficient mice are protected against tobacco-induced inflammation and increased emphysema. Am J Physiol Lung Cell Mol Physiol 2010;299(6):L834-42

Expert Opin. Drug Discov. (2014) 9(6)

133. Ajiro Y, Tokuhashi Y, Matsuzaki H, et al. Impact of passive smoking on the bones of rats. Orthopedics 2010;33(2):90-5 134. Glynos C, Dupont LL, Vassilakopoulos T, et al. The role of soluble guanylyl cyclase in chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2013;188(7):789-99 135. Schweitzer KS, Johnstone BH, Garrison J, et al. Adipose stem cell treatment in mice attenuates lung and systemic injury induced by cigarette smoking. Am J Respir Crit Care Med 2011;183(2):215-25

Animal models of COPD

136.

Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Queensland on 05/26/14 For personal use only.

.

137.

.

138.

Marwick JA, Caramori G, Stevenson CS, et al. Inhibition of PI3Kdelta restores glucocorticoid function in smokinginduced airway inflammation in mice. Am J Respir Crit Care Med 2009;179(7):542-8 The authors demonstrate a novel therapeutic approach whereby inhibition of phosphatidylinositol 3-kinase d prevents CS-induced suppression of histone deacetylase 2 activity and restores sensitivity of CS-induced inflammation to glucocorticoids. Bozinovski S, Uddin M, Vlahos R, et al. Serum amyloid A opposes lipoxin A(4) to mediate glucocorticoid refractory lung inflammation in chronic obstructive pulmonary disease. Proc Natl Acad Sci USA 2012;17;109(3):935-40 Implication of serum amyloid A as a mediator of steroid-resistant neutrophilic inflammation in humans and mouse. Hakim A, Adcock IM, Usmani OS. Corticosteroid resistance and novel antiinflammatory therapies in chronic

obstructive pulmonary disease: current evidence and future direction. Drugs 2012;72(10):1299-312 139.

Nie YC, Wu H, Li PB, et al. Characteristic comparison of three rat models induced by cigarette smoke or combined with LPS: to establish a suitable model for study of airway mucus hypersecretion in chronic obstructive pulmonary disease. Pulm Pharmacol Ther 2012;25(5):349-56

140.

D’Hulst A I, Vermaelen KY, Brusselle GG, et al. Time course of cigarette smoke-induced pulmonary inflammation in mice. Eur Respir J 2005;26(2):204-13

141.

Bracke KR, Verhamme FM, Seys LJ, et al. Role of CXCL13 in cigarette smoke-induced lymphoid follicle formation and chronic obstructive pulmonary disease. Am J Respir Crit Care Med 2013;188(3):343-55

142.

van der Deen M, Timens W, Timmer-Bosscha H, et al. Reduced inflammatory response in cigarette smoke

Expert Opin. Drug Discov. (2014) 9(6)

exposed Mrp1/Mdr1a/1b deficient mice. Respir Res 2007;8:49 143. Christensen PJ, Preston AM, Ling T, et al. Pneumocystis murina infection and cigarette smoke exposure interact to cause increased organism burden, development of airspace enlargement, and pulmonary inflammation in mice. Infect Immun 2008;76(8):3481-90

Affiliation Michael Fricker1 PhD, Andrew Deane1 MSc & Philip M Hansbro†2 PhD † Author for correspondence 1 University of Newcastle and Hunter Medical Research Institute, Priority Research Centre for Asthma and Respiratory Disease, New Lambton Heights, New South Wales, Australia 2 Professor, University of Newcastle and Hunter Medical Research Institute, Priority Research Centre for Asthma and Respiratory Disease, New Lambton Heights, New South Wales, Australia E-mail: [email protected]

645

Animal models of chronic obstructive pulmonary disease.

Chronic obstructive pulmonary disease (COPD) is a leading global cause of mortality and chronic morbidity. Inhalation of cigarette smoke is the princi...
494KB Sizes 4 Downloads 10 Views