Journal Information
Vol. 51. Issue 3.
Pages 121-127 (March 2015)
Visits
13296
Vol. 51. Issue 3.
Pages 121-127 (March 2015)
Review
Full text access
Animal Models of Chronic Obstructive Pulmonary Disease
Modelos animales de enfermedad pulmonar obstructiva crónica
Visits
13296
Sandra Pérez-Rial, Álvaro Girón-Martínez, Germán Peces-Barba
Corresponding author
gpeces@fjd.es

Corresponding author.
Laboratorio de Neumología, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES-UAM, Madrid, Spain
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (2)
Tables (3)
Table 1. Examples of Cigarette Smoke-Induced COPD.
Table 2. Strains of Natural Mutant Mice That Develop Emphysema.
Table 3. Examples of “Knockout” Models of Emphysematous Mice.
Show moreShow less
Abstract

Animal models of disease have always been welcomed by the scientific community because they provide an approach to the investigation of certain aspects of the disease in question.

Animal models of COPD cannot reproduce the heterogeneity of the disease and usually only manage to represent the disease in its milder stages. Moreover, airflow obstruction, the variable that determines patient diagnosis, not always taken into account in the models. For this reason, models have focused on the development of emphysema, easily detectable by lung morphometry, and have disregarded other components of the disease, such as airway injury or associated vascular changes.

Continuous, long-term exposure to cigarette smoke is considered the main risk factor for this disease, justifying the fact that the cigarette smoke exposure model is the most widely used. Some variations on this basic model, related to exposure time, the association of other inducers or inhibitors, exacerbations or the use of transgenic animals to facilitate the identification of pathogenic pathways have been developed. Some variations or heterogeneity of this disease, then, can be reproduced and models can be designed for resolving researchers’ questions on disease identification or treatment responses.

Keywords:
Chronic obstructive pulmonary disease
Animal model
Smoking
Emphysema
Transgenic animals
Exacerbation
Autoimmune
Therapeutic assays
Resumen

El desarrollo de modelos animales de una enfermedad ha sido siempre bien acogido por la comunidad científica porque permite realizar una aproximación a la investigación de determinados aspectos de la misma.

Los modelos animales de la EPOC no pueden llegar a reproducir la heterogeneidad de esta enfermedad y generalmente solo llegan a representar los estadios más leves de la misma. Además, la obstrucción al flujo aéreo, variable que determina el diagnóstico en un paciente, no siempre se tiene en cuenta en los modelos. Por este motivo, los modelos se han centrado en el desarrollo de enfisema, fácilmente detectable por morfometría pulmonar, sin prestar atención a otros componentes de la enfermedad, como la lesión de las vías aéreas o las alteraciones vasculares asociadas.

La exposición continua y prolongada al humo de tabaco se considera el principal factor de riesgo de esta enfermedad, lo que justifica que sea el modelo de exposición al humo de tabaco el más ampliamente utilizado. Sobre esta base de modelo podemos encontrar algunas variantes relacionadas con el tiempo de exposición, la asociación de otros inductores o inhibidores, las exacerbaciones o el uso de animales transgénicos que facilitan la identificación de las vías patogénicas. Es posible, por tanto, reproducir algunas variantes o heterogeneidades de esta enfermedad y diseñar uno u otro modelo que sea capaz de responder a una u otra pregunta de investigación, dirigida bien a una identificación patogénica y/o bien a una respuesta terapéutica.

Palabras clave:
Enfermedad pulmonar obstructiva crónica
Modelo animal
Tabaco
Enfisema
Transgénico
Exacerbación
Autoinmune
Ensayos terapéuticos
Full Text
Introduction

Chronic obstructive pulmonary disease (COPD) has a huge global impact, and clinicians must make use of all the tools available to tackle the many aspects of this disease. The development of animal models can help address problems such as under-diagnosis, frail exacerbator patients, and existing uncertainties about the development of one or other clinical form of the disease or its natural history (in which cases with minor and accelerated disease progression are mixed). Similarly, all new therapeutic trials are generally based on an earlier study in an animal model.

Smoking is the leading cause of COPD, but its ability to generate a permanent inflammatory response depends on the patient's susceptibility. For this reason, animal models of COPD developed by exposure to cigarette smoke are primarily chosen to study the pathogenic mechanisms of the disease and of susceptibility to development and progression. In these cases, the use of transgenic animals, in which a particular metabolic pathway is inhibited or activated, helps researchers understand the pathogenic pathways that exist in each case. Likewise, new approaches to the classification of COPD proposed by both the GOLD initiative1 and Spanish COPD guidelines (GesEPOC)2 place greater emphasis on exacerbations due to their effect on the severity of symptoms, progression of the obstruction, and mortality. For this reason, interest has grown in the study of exacerbations in models of COPD, and the results may help to improve knowledge of the mechanisms underlying the condition in the exacerbator patient.

When evaluating the results of studies in animal models of any disease, the limitation of having to extrapolate a conclusion as to what is potentially present in a patient must always be taken into consideration. However, they are an essential part of clinical research when used as “preclinical models”, an increasingly widespread term that encompasses the notion of translation into clinical practice that must form the basis of any study design.

Models of Cigarette Smoke-Induced COPD

Models of cigarette smoke-induced COPD are those that best reflect the inflammatory and pathogenic mechanisms of the disease and, consequently, those that are potentially better suited to testing new therapies. Exposure to cigarette smoke has been applied in numerous animal species, such as dogs, guinea pigs, rabbits, rats, and mice. Of these, guinea pigs and mice have proven to be most susceptible to the development of COPD through prolonged exposure.3 There are two general procedures for administering cigarette smoke: the so-called “nose only” method, where the smoke is channeled directly into the animal's nose, and “whole body” administration (Fig. 1), where the animal is placed in a chamber filled with a controlled concentration of smoke to ensure complete exposure to stable, non-toxic carboxyhemoglobin levels.4,5 Conceptually different, both methods have been widely used, and have shown similar findings as regards the presence of inflammatory cell populations, cytokine levels, changes in lung remodeling, and therapeutic response6 (Fig. 2).

Fig. 1.

Whole body tobacco exposure system.

(0.14MB).
Fig. 2.

Histological sections (H&E) of the lungs of mice exposed to ambient air (A) and mice exposed to tobacco smoke for 6 months (B), showing emphysema.

(0.28MB).

In approximately 90% of patients, COPD is caused by smoking an average of at least 10 pack-years, and they develop a disease that can take different clinical forms, with different levels of progression and severity.7 Animal models of COPD, guinea pig or murine are usually established over a 6-month exposure period,8 although major inflammatory and morphometric changes can already be detected after the second month.9 They do not usually reach the stage equivalent to severe COPD in a patient, but they can develop many of the characteristics typical of this disease, such as chronic inflammation with increased neutrophil and macrophage counts, presence of CD4 and CD8T lymphocytes, mucus hypersecretion, changes in lung function, emphysema, and vascular and airway remodeling.10

The murine model of cigarette smoke exposure is the most widely used, due to its low cost and easy management, well-mapped genome, the availability of many transgenic variants, a wide range of specific antibodies for laboratory use, and a large number of strains with differing susceptibilities to cigarette smoke. Strain-dependent susceptibility for developing COPD is well identified in the murine model,11–14 and the pulmonary morphometric pattern of COPD can be generated when mice are exposed to cigarette smoke for at least 3 to 6 months, with typical inflammatory cells, inflammatory mediators and functional changes characteristic of the disease.15

Guinea pigs are also a good choice for generating models of COPD, as these animals are very susceptible to developing the disease after only a few months of exposure.16 In 1990, Wright and Churg published one of the first studies in guinea pigs exposed to cigarette smoke.17 In this case, after 12 months of exposure the guinea pigs developed emphysema and presented lung function changes very similar to those found in smokers with COPD. The main difficulty in the use of this species comes from the limited availability of specific antibodies. Rats, the species of rodent closest to mice and guinea pigs, are rarely used as models of COPD because they are more resistant to developing changes due to cigarette smoke exposure.18Table 1 summarizes some relevant findings described in this model.

Table 1.

Examples of Cigarette Smoke-Induced COPD.

Animal model  Methodology  Finding  Reference 
Serpin B1 KO mice  Chronic exposure  Not associated with severe emphysema  83 
NZWLac/J, AJ, SJL, C57BL/6, and AKR mice  Chronic exposure  The degree of susceptibility to developing lung injury is strain-dependent  12 
NZWLac/J and AKR mice  Chronic exposure  Egr-1 (proinflammatory marker) greatly increased in the susceptible AKR strain, but far less so in the NZWLac/J strain  84 
C57BL/6 (CD8−/−) mice  Chronic exposure  CD8+ T lymphocytes are essential for the development of emphysema  85 
C57BL/6 mice  Acute exposure  Increased markers of DNA damage by oxidative stress (8-OHdG and 4-HNE)  86 
ICR (Nrf2−/−) mice  Chronic exposure  More susceptible to developing emphysema, with increased antioxidant enzyme expression  78 
Guinea pig  Chronic exposure  Increase in vascular remodeling and arterial pulmonary pressure  87 
C57BL/6 and “pallid” mice  Chronic exposure  The α1-antitrypsin levels determine the emphysematous profile  88 
Guinea pig  Chronic exposure  Role of MMPs in the development of emphysema and airway remodeling  89 
C57BL/6 mice  Acute exposure (direct and indirect)  The intensity of the inflammatory response depends on the composition of the cigarette smoke  90 
C57BL/6 mice (sGCα1(−/−)Acute and chronic exposure  Reduction in sGC expression contributing to airflow limitation  91 
Mice  Sub-chronic exposure  Anti-inflammatory effect of PPAR-γ ligands  92 
Mice  Chronic exposure  The neutralization of CXCL13 partially blocks the inflammatory response and alveolar wall destruction  93 
Mice  Chronic exposure  Blocking T lymphocytes may be effective as therapy for COPD  94 
C57BL/6 and BALB/cJ mice  Chronic exposure  Cigarette smoke triggers an antigen-dependent response in which CD4+ and CD8+ lymphocytes participate  95 
C57BL/6 and 129S2/SvHsd mice  Acute exposure  Implication of the pro-inflammatory monocytes in susceptibility to developing lung injury  5 

Models of COPD due to chronic cigarette smoke exposure continue to be limited insofar as they are unable to reproduce some of the characteristics of this complex, heterogeneous disease. So far, attempts to develop known clinical phenotypes, such as the exacerbator or accelerated progression types, or forms with bacterial colonization, for example, have failed. Some studies, however, have attempted to address these questions by combining agents. Exposure to toxic and irritant gases such as nitrogen dioxide, ozone or sulfur dioxide causes more severe lung damage than cigarette smoke.19–22

Models of COPD Exacerbations

Exacerbations are a characteristic of COPD that, if repeated, determine the poor clinical course of the patient, as they are associated with greater disease progression, poorer quality of life and higher risk of mortality. The availability of animal models of exacerbation gives researchers the chance to study associated pathogenic mechanisms and detect possible associated biological markers.

Most infectious COPD exacerbations are viral in origin (75%), while the remainder are bacterial. Studies in in vivo models have demonstrated the effect of viral infection on mice previously exposed, both short- and long-term, to cigarette smoke. Inflammation of the lung is more severe if the viral infection affects an animal previously exposed to cigarette smoke, and also accelerates emphysema progression and the severity of airway damage.23,24

The most commonly isolated bacterium in COPD exacerbations is nontypeable Haemophilus influenzae (NTHI). For this reason, the results obtained in models of this infection in healthy rats25 vs. those previously exposed to cigarette smoke are particularly interesting. After C57BL/6 mice had been exposed to cigarette smoke for 8 weeks, NTHI infection caused a more severe inflammatory response and greater lung damage than in previously healthy animals.26,27

Bacterial lipopolysaccharides (LPS) have been used alone, in long-term administration,28 or in combination with short periods of exposure to cigarette smoke29 to develop models of emphysema. However, single massive insult can cause an inflammatory response that is accompanied by fever, mucus hypersecretion and bronchoconstriction, which reproduces symptoms of an exacerbation30 seen on computed tomography.31

Models of Severe COPD by Combining Induction Agents

In the more severe stages of COPD a clear breakdown of the lung “maintenance program” occurs that can inevitably lead to emphysema and pulmonary hypertension. There are various models of “frail” (very severe) COPD pathology, such as the combination of cigarette smoke exposure and vascular endothelial growth factor (VEGF) inhibitor.32 Exposure to cigarette smoke causes a significant decrease in VEGF and VEGF receptor-2 (VEGFR-2) expression in animal models of emphysema. Furthermore, treatment with the VEGF receptor blocker SU5416 induces alveolar cell apoptosis, capillary retraction, and alveolar space enlargement.33 For this reason, emphysema presents as a VEGF deficiency that compromises the survival of the endothelial cells and consequently the lung's maintenance program. Other results can be achieved by combining cigarette smoke exposure and hypoxia induction. This model can lead to pulmonary hypertension, a condition present only in advanced severe COPD.34

Transgenic Models of COPD

Before the advent of targeted genetic engineering, some mutant strains of C57BL/6 mice that spontaneously developed emphysema had appeared (Table 2). These were the “blotchy” mice that have an abnormal translation of the Menkes gene on the X chromosome,35 causing defects in lung connective tissue proteins, which affects the structure and function of the lungs, causing emphysema36; “Tight Skin” mice, with a mutation in fibrillin-1, one of the key components of the microfibrils in the lung extracellular matrix, which causes oxidative stress and cell death, injury cascades central to the development of emphysema37,38; “Beige” mice, in which the lungs appear normal at birth but, due to the deletion of Lyst, do not form alveoli normally during development39; “Pallid” mice,40 with a mutation that affects syntaxin-13 (a cell membrane protein), resulting in the gradual and progressive development of emphysema41; and more recently, “Osteopetrotic” mice, which are macrophage colony-stimulating factor-deficient and eventually develop emphysema.42 One of the major technological breakthroughs of the last few decades has been the development of transgenic animals. These are animals in which a gene that does not form part of their genome, and which will sequence a certain pathway of interest, is inserted by intranuclear injection in the early embryonic phases.13,15,43–45 One of the first applications of transgenic technology to COPD was the constitutive overexpression of human collagenase-1 (MMP-1) in mice, which causes emphysema46 by degradation of type III collagen in the alveolar walls.47 The constitutive expression of transgenes, however, does not distinguish the lung's own development process. To overcome this, the transgenic expression construct was developed. Thus, overexpression of IL-13,48 a cytokine produced by T-helper type 2 (Th2) lymphocytes, or overexpression of IFN-γ,49 the main product of T-helper type 1 (Th1) lymphocytes, are two important examples of inducible conditional transgenes. In the case of IL-13 transgenic mice (“Dutch”), this leads to MMP-9 and MMP-12-dependent emphysema in adult mice.50 In these animals, IL-13 is overexpressed only when they are exposed to tetracycline, thereby allowing investigators to activate overexpression after the lung is fully developed. MMP-9-mediated activation of TGF-β appears to be responsible for collagen remodeling in this model. However, in IFN-γ transgenic mice (“British”), the inflammatory component appears to be more subtle, with prominent apoptosis but no associated airways disease. These are only two examples that demonstrate the complexity of inflammatory networks, and how unexpected findings in animal models have led to the search for new mediators in human disease. Other studies show how TNF-α induction in the adult lung facilitates the formation of lymphoid tissue and emphysema, providing a model for research into the pathogenic effects of TNF-α in the lung,51 or how prothymosin-α (ProT-α) expression contributes to the pathogenesis of emphysema by increasing acetylation of histones and expression of NF-κB-dependent MMP-2 and MMP-9, especially after cigarette smoke exposure.52

Table 2.

Strains of Natural Mutant Mice That Develop Emphysema.

Mouse  Mutation  Pulmonary phenotype  Reference 
Blotchy (Blo)Abnormal translation in the Menkes gene on the X chromosomeAir space enlargement  35,36 
Disorganization of the elastic fibers  37,38 
Tight skin (Tsk+/−)  Duplication of fibrillin-1  Abnormal air space development, with development of panlobular emphysema  39 
Beige (BgDeletion in Lyst  Normal at birth, but with abnormal neonatal alveolization  40,41 
Pallid (PaSyntaxin-3  Develop mild, late onset emphysema  42 
Osteopetrotic (OpMacrophage colony-stimulating factor deficiency  Reduced number of alveolar macrophages and emphysema   

An alternative to the transgenic model is the “knockout” model (Table 3), in which the expression of a certain gene is inhibited, thereby enabling the function of proteins dependent on this gene to be determined. Sometimes, gene inhibition protects against the development of emphysema, as in the case of inhibition of MMP-12 expression, which impairs alveolar macrophage recruitment and thereby protects against the development of emphysema.53 Another example is the absence of neutrophil elastase (NE), which also appears to protect against the development of smoke-induced emphysema.54 In both cases, the direct role of these proteins in emphysema has been demonstrated, highlighting the interdependence of the proteinases and inflammatory cells that mediate lung destruction in response to cigarette smoke. At other times, the deletion interferes with alveogenesis. In this case, platelet-derived growth factor A (PDGF-A)-deficient mice develop emphysema due to loss of myofibroblasts and the associated elastin fiber deposits.55 Double knockout mice for fibroblast growth factor receptors 3 and 4 (FGFR-3 and -4) have abnormal alveolar formation and septation,56 while elastin-deficient mice have fewer dilated distal air sacs and arrested airway development.57 In other cases, deletion of certain genes causes alveolar space enlargement. This is true of integrin αVβ6-deficient mice, in which TGF-β activation in alveolar air spaces does not occur, leading to development of MMP-12-dependent emphysema.58 Other examples are knockout mice for pulmonary surfactant protein D (SP-D), which present macrophage activation, production of MMPs and air space enlargement,59 or tissue inhibitor of metalloproteinase-3 (TIMP-3) deficiency, which appears to combine air space enlargement with the gradual development of emphysema.60

Table 3.

Examples of “Knockout” Models of Emphysematous Mice.

Gene  Pulmonary phenotype  Reference 
Macrophage elastase (MMP-12)  Complete protection against the development of emphysema after exposure to cigarette smoke  53 
Neutrophil elastase (NE)  Protection against cigarette smoke-induced emphysema  54 
Elastin  Fewer dilated distal air sacs and arrested airway development  57 
Platelet-derived growth factor A (PDGF-A)  Lack of tropoelastin and failure in alveolar septation  55 
Fibroblast growth factor receptor (FGFR-3 and -4)  Abnormal alveolar formation and septation  56 
Integrin αvβ6  Spontaneous development of MMP-12-dependent emphysema  58 
Pulmonary surfactant protein D (SP-D)  Macrophage activation, production of MMPs and subsequent emphysema  59 
Tissue inhibitor of metalloprotease-3 (TIMP-3)  Air space enlargement with gradual development of emphysema  60 

To overcome the cross-species barrier in the murine model, certain murine genes can be eliminated and human genes inserted (“knocked in”) under the control of murine promoters. Emphysema-prone mice, in which the murine alpha-1 antitrypsin (A1AT) genes have been removed and replaced by normal or deficient human A1AT genes, have also been developed.61

Autoimmune Models of COPD

Pulmonary inflammation in severe COPD involves a large number of activated Th1T lymphocytes, B lymphocytes and CD8 lymphocytes, which persist for years, even after smoking cessation; this is consistent with a self-perpetuating process, which is one of the characteristics of autoimmune diseases. This chain of events suggests that the adaptive immune response in COPD, together with its persistence after smoking cessation, could be due to a response to autoantigens. Initially, this was merely a hypothesis,62–64 but since then new evidence, including the development of the first animal model of autoimmune emphysema,66–68 would seem to confirm the suggestion.65 The presence of anti-elastin autoantibodies69 and other autoantigens70,71 has been correlated with emphysema severity, and induction of autoantibodies against lung matrix proteins has been shown to increase the smoke-induced immune response in mice previously immunized with a mixture of lung extracellular matrix proteins.72

Models for Therapeutic Trials in COPD

Current treatments do little to inhibit chronic inflammation, do not reverse COPD pathology, and do not modify the factors that initiate and lead to disease progression in the long term. It is clear, therefore, that new therapies that can prevent COPD induction and progression must be developed, and this is only possible through animal models that accurately reflect the physiopathology of the disease. Many anti-COPD drugs in clinical development have been identified from studies in animal models. Various inhibitors of inflammatory mediators are being developed for the treatment of COPD, although to date the results of tests using LTB4, TNF-α, IL-1, IL-8, and EGF inhibitors have been disappointing.73 Studies in animals exposed to cigarette smoke and treated with synthetic neutrophil elastase inhibitors have shown their potential anti-inflammatory activity.74 Similarly, findings in animal models of cigarette smoke-induced airway inflammation support the potential therapeutic usefulness of kinase inhibitors (p38 MAPK and PI3K) in COPD.75 The antioxidant enzyme Gpx-1 protects against lung inflammation and cigarette smoke-induced emphysema in mice,76 and a Gpx mimetic also reduced lung inflammation when administered both prophylactically and therapeutically.76,77 In other studies, deletion of the Nrf2 antioxidant stress response gene led to increased lung inflammation and emphysema in mice exposed to cigarette smoke,78 and an Nrf2 activator is currently undergoing clinical trials for COPD.73

New COPD drugs that can reduce the rate of pulmonary destruction and airflow limitation, and even arrest or reverse the underlying processes have yet to be discovered. In this regard, some evidence suggests that retinoic acid significantly slows elastase-induced emphysema in rats,79 and this has sparked interest in the retinoids80 and other growth factors as potential therapeutic agents,81 although how these can be used has yet to be confirmed. Studies published by our group show that liver growth factor (LGF) has pulmonary anti-fibrotic activity, and can improve lung function and partially revert the deposit of matrix proteins following CdCl2 administration in a model of fibrosis in rats.82

In conclusion, studies in animal models of COPD continue to contribute important information. These valuable tools further our understanding of the pathogenic aspects of the disease and help develop therapeutic clinical trials. The inherent heterogeneity of the disease can also be reflected in animal models developed by using different combinations or doses of induction agents. This is why it is important to choose the model according to whether the research is focused on pathogenesis, diagnosis or treatment.

Conflict of Interests

The authors declare that they have no conflict of interests.

References
[1]
A. Lopez-Giraldo, R. Rodriguez-Roisin, A. Agusti.
Chronic obstructive pulmonary disease: the golden decade. Implications for the diagnosis, prevention and treatment of chronic obstructive pulmonary disease.
[2]
M. Miravitlles, J.J. Soler-Cataluna, M. Calle, J. Molina, P. Almagro, J.A. Quintano, et al.
Spanish guideline for COPD (GesEPOC).
Arch Bronconeumol, 50 (2014), pp. 1-16
[3]
G. Peces-Barba, S. Heili.
Modelos animales de EPOC.
Arch Bronconeumol, 43 (2007), pp. 30-37
[4]
S. Perez-Rial, L. Del Puerto-Nevado, N. Gonzalez-Mangado, G. Peces-Barba.
Early detection of susceptibility to acute lung inflammation by molecular imaging in mice exposed to cigarette smoke.
Mol Imaging, 10 (2011), pp. 398-405
[5]
S. Perez-Rial, L. del Puerto-Nevado, R. Terron-Exposito, A. Giron-Martinez, N. Gonzalez-Mangado, G. Peces-Barba.
Role of recently migrated monocytes in cigarette smoke-induced lung inflammation in different strain of mice.
[6]
M. Leberl, A. Kratzer, L. Taraseviciene-Stewart.
Tobacco smoke induced COPD/emphysema in the animal model-are we all on the same page?.
Front Physiol, 4 (2013), pp. 91
[7]
M. Miravitlles, J.J. Soler-Cataluna, M. Calle, J. Molina, P. Almagro, J. Antonio Quintano, et al.
Spanish Guideline for COPD (GesEPOC).
Arch Bronconeumol, 50 (2014), pp. 1-16
[8]
J.L. Wright, M. Cosio, A. Churg.
Animal models of chronic obstructive pulmonary disease.
Am J Physiol Lung Cell Mol Physiol, 295 (2008), pp. L1-L15
[9]
B.W. van der Strate, D.S. Postma, C.A. Brandsma, B.N. Melgert, M.A. Luinge, M. Geerlings, et al.
Cigarette smoke-induced emphysema: a role for the B cell?.
Am J Respir Crit Care Med, 173 (2006), pp. 751-758
[10]
R. Vlahos, S. Bozinovski.
Recent advances in pre-clinical mouse models of COPD.
Clin Sci (Lond), 126 (2014), pp. 253-265
[11]
T.H. March, E.B. Barr, G.L. Finch, F.F. Hahn, C.H. Hobbs, M.G. Menache, et al.
Cigarette smoke exposure produces more evidence of emphysema in B6C3F1 mice than in F344 rats.
Toxicol Sci, 51 (1999), pp. 289-299
[12]
A. Guerassimov, Y. Hoshino, Y. Takubo, A. Turcotte, M. Yamamoto, H. Ghezzo, et al.
The development of emphysema in cigarette smoke-exposed mice is strain dependent.
Am J Respir Crit Care Med, 170 (2004), pp. 974-980
[13]
G.G. Brusselle, K.R. Bracke, T. Maes, A.I. D’Hulst, K.B. Moerloose, G.F. Joos, et al.
Murine models of COPD.
Pulm Pharmacol Ther, 19 (2006), pp. 155-165
[14]
B. Bartalesi, E. Cavarra, S. Fineschi, M. Lucattelli, B. Lunghi, P.A. Martorana, et al.
Different lung responses to cigarette smoke in two strains of mice sensitive to oxidants.
Eur Respir J, 25 (2005), pp. 15-22
[15]
S.D. Shapiro.
Animal models for chronic obstructive pulmonary disease: age of klotho and marlboro mice.
Am J Respir Cell Mol Biol, 22 (2000), pp. 4-7
[16]
J.L. Wright, A. Churg.
A model of tobacco smoke-induced airflow obstruction in the guinea pig.
Chest, 121 (2002), pp. 188S-191S
[17]
J.L. Wright, A. Churg.
Cigarette smoke causes physiologic and morphologic changes of emphysema in the guinea pig.
Am Rev Respir Dis, 142 (1990), pp. 1422-1428
[18]
C.S. Stevenson, C. Docx, R. Webster, C. Battram, D. Hynx, J. Giddings, et al.
Comprehensive gene expression profiling of rat lung reveals distinct acute and chronic responses to cigarette smoke inhalation.
Am J Physiol Lung Cell Mol Physiol, 293 (2007), pp. L1183-L1193
[19]
A. Farone, S. Huang, J. Paulauskis, L. Kobzik.
Airway neutrophilia and chemokine mRNA expression in sulfur dioxide-induced bronchitis.
Am J Respir Cell Mol Biol, 12 (1995), pp. 345-350
[20]
S. Shore, L. Kobzik, N.C. Long, W. Skornik, C.J. Van Staden, L. Boulet, et al.
Increased airway responsiveness to inhaled methacholine in a rat model of chronic bronchitis.
Am J Respir Crit Care Med, 151 (1995), pp. 1931-1938
[21]
P. Chitano, J.J. Hosselet, C.E. Mapp, L.M. Fabbri.
Effect of oxidant air pollutants on the respiratory system: insights from experimental animal research.
Eur Respir J, 8 (1995), pp. 1357-1371
[22]
M. Wegmann, H. Renz, U. Herz.
Long-term NO2 exposure induces pulmonary inflammation and progressive development of airflow obstruction in C57BL/6 mice: a mouse model for chronic obstructive pulmonary disease?.
Pathobiology, 70 (2002), pp. 284-286
[23]
R.C. Gualano, M.J. Hansen, R. Vlahos, J.E. Jones, R.A. Park-Jones, G. Deliyannis, et al.
Cigarette smoke worsens lung inflammation and impairs resolution of influenza infection in mice.
Respir Res, 9 (2008), pp. 53
[24]
M.J. Kang, C.G. Lee, J.Y. Lee, C.S. Dela Cruz, Z.J. Chen, R. Enelow, et al.
Cigarette smoke selectively enhances viral PAMP- and virus-induced pulmonary innate immune and remodeling responses in mice.
J Clin Invest, 118 (2008), pp. 2771-2784
[25]
P. Morey, C. Viadas, B. Euba, D.W. Hood, M. Barberan, C. Gil, et al.
Relative contributions of lipooligosaccharide inner and outer core modifications to nontypeable Haemophilus influenzae pathogenesis.
Infect Immun, 81 (2014), pp. 4100-4111
[26]
G.J. Gaschler, M. Skrtic, C.C. Zavitz, M. Lindahl, P.O. Onnervik, T.F. Murphy, et al.
Bacteria challenge in smoke-exposed mice exacerbates inflammation and skews the inflammatory profile.
Am J Respir Crit Care Med, 179 (2009), pp. 666-675
[27]
G.J. Gaschler, C.C. Zavitz, C.M. Bauer, M.R. Stampfli.
Mechanisms of clearance of nontypeable Haemophilus influenzae from cigarette smoke-exposed mouse lungs.
Eur Respir J, 36 (2010), pp. 1131-1142
[28]
J. Stolk, A. Rudolphus, P. Davies, D. Osinga, J.H. Dijkman, L. Agarwal, et al.
Induction of emphysema and bronchial mucus cell hyperplasia by intratracheal instillation of lipopolysaccharide in the hamster.
J Pathol, 167 (1992), pp. 349-356
[29]
Y.C. Nie, H. Wu, P.B. Li, Y.L. Luo, C.C. Zhang, J.G. Shen, 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, 25 (2012), pp. 349-356
[30]
J. Spond, M.M. Billah, R.W. Chapman, R.W. Egan, J.A. Hey, A. House, et al.
The role of neutrophils in LPS-induced changes in pulmonary function in conscious rats.
Pulm Pharmacol Ther, 17 (2004), pp. 133-140
[31]
S. Kobayashi, R. Fujinawa, F. Ota, T. Angata, M. Ueno, T. Maeno, et al.
A single dose of lipopolysaccharide into mice with emphysema mimics human chronic obstructive pulmonary disease exacerbation as assessed by micro-computed tomography.
Am J Respir Cell Mol Biol, 49 (2014), pp. 971-977
[32]
I. Edirisinghe, S.R. Yang, H. Yao, S. Rajendrasozhan, S. Caito, D. Adenuga, et al.
VEGFR-2 inhibition augments cigarette smoke-induced oxidative stress and inflammatory responses leading to endothelial dysfunction.
FASEB J, 22 (2008), pp. 2297-2310
[33]
Y. Kasahara, R.M. Tuder, L. Taraseviciene-Stewart, T.D. Le Cras, S. Abman, P.K. Hirth, et al.
Inhibition of VEGF receptors causes lung cell apoptosis and emphysema.
J Clin Invest, 106 (2000), pp. 1311-1319
[34]
E. Olea, E. Ferrer, J. Prieto-Lloret, C. Gonzalez-Martin, V. Vega-Agapito, E. Gonzalez-Obeso, et al.
Effects of cigarette smoke and chronic hypoxia on airways remodeling and resistance. Clinical significance.
Respir Physiol Neurobiol, 179 (2014), pp. 305-313
[35]
J.F. Mercer, A. Grimes, L. Ambrosini, P. Lockhart, J.A. Paynter, H. Dierick, et al.
Mutations in the murine homologue of the Menkes gene in dappled and blotchy mice.
Nat Genet, 6 (1994), pp. 374-378
[36]
D.E. Fisk, C. Kuhn.
Emphysema-like changes in the lungs of the blotchy mouse.
Am Rev Respir Dis, 113 (1976), pp. 787-797
[37]
C.M. Kielty, M. Raghunath, L.D. Siracusa, M.J. Sherratt, R. Peters, C.A. Shuttleworth, et al.
The Tight skin mouse: demonstration of mutant fibrillin-1 production and assembly into abnormal microfibrils.
J Cell Biol, 140 (1998), pp. 1159-1166
[38]
M. Podowski, C.L. Calvi, C. Cheadle, R.M. Tuder, S. Biswals, E.R. Neptune.
Complex integration of matrix, oxidative stress, and apoptosis in genetic emphysema.
Am J Pathol, 175 (2009), pp. 84-96
[39]
B. Starcher, I. Williams.
The beige mouse: role of neutrophil elastase in the development of pulmonary emphysema.
Exp Lung Res, 15 (1989), pp. 785-800
[40]
P.A. Martorana, T. Brand, C. Gardi, P. van Even, M.M. de Santi, P. Calzoni, et al.
The pallid mouse. A model of genetic alpha 1-antitrypsin deficiency.
Lab Invest, 68 (1993), pp. 233-241
[41]
L. Huang, Y.M. Kuo, J. Gitschier.
The pallid gene encodes a novel, syntaxin 13-interacting protein involved in platelet storage pool deficiency.
Nat Genet, 23 (1999), pp. 329-332
[42]
Y. Shibata, Z. Zsengeller, K. Otake, N. Palaniyar, B.C. Trapnell.
Alveolar macrophage deficiency in osteopetrotic mice deficient in macrophage colony-stimulating factor is spontaneously corrected with age and associated with matrix metalloproteinase expression and emphysema.
Blood, 98 (2001), pp. 2845-2852
[43]
R. Mahadeva, S.D. Shapiro.
Chronic obstructive pulmonary disease * 3: experimental animal models of pulmonary emphysema.
Thorax, 57 (2002), pp. 908-914
[44]
R.M. Tuder, S. McGrath, E. Neptune.
The pathobiological mechanisms of emphysema models: what do they have in common?.
Pulm Pharmacol Ther, 16 (2003), pp. 67-78
[45]
R. Vlahos, S. Bozinovski, R.C. Gualano, M. Ernst, G.P. Anderson.
Modelling COPD in mice.
Pulm Pharmacol Ther, 19 (2006), pp. 12-17
[46]
J. D’Armiento, S.S. Dalal, Y. Okada, R.A. Berg, K. Chada.
Collagenase expression in the lungs of transgenic mice causes pulmonary emphysema.
Cell, 71 (1992), pp. 955-961
[47]
T. Shiomi, Y. Okada, R. Foronjy, J. Schiltz, R. Jaenish, S. Krane, et al.
Emphysematous changes are caused by degradation of type III collagen in transgenic mice expressing MMP-1.
Exp Lung Res, 29 (2003), pp. 1-15
[48]
T. Zheng, Z. Zhu, Z. Wang, R.J. Homer, B. Ma, R.J. Riese Jr., et al.
Inducible targeting of IL-13 to the adult lung causes matrix metalloproteinase- and cathepsin-dependent emphysema.
J Clin Invest, 106 (2000), pp. 1081-1093
[49]
Z. Wang, T. Zheng, Z. Zhu, R.J. Homer, R.J. Riese, H.A. Chapman Jr., et al.
Interferon gamma induction of pulmonary emphysema in the adult murine lung.
J Exp Med, 192 (2000), pp. 1587-1600
[50]
J.A. Elias, M.J. Kang, K. Crothers, R. Homer, C.G. Lee.
State of the art. Mechanistic heterogeneity in chronic obstructive pulmonary disease: insights from transgenic mice.
Proc Am Thorac Soc, 3 (2006), pp. 494-498
[51]
B.R. Vuillemenot, J.F. Rodriguez, G.W. Hoyle.
Lymphoid tissue and emphysema in the lungs of transgenic mice inducibly expressing tumor necrosis factor-alpha.
Am J Respir Cell Mol Biol, 30 (2004), pp. 438-448
[52]
B.H. Su, Y.L. Tseng, G.S. Shieh, Y.C. Chen, Y.C. Shiang, P. Wu, et al.
Prothymosin alpha overexpression contributes to the development of pulmonary emphysema.
Nat Commun, 4 (2013), pp. 1906
[53]
R.D. Hautamaki, D.K. Kobayashi, R.M. Senior, S.D. Shapiro.
Requirement for macrophage elastase for cigarette smoke-induced emphysema in mice.
Science, 277 (1997), pp. 2002-2004
[54]
S.D. Shapiro, N.M. Goldstein, A.M. Houghton, D.K. Kobayashi, D. Kelley, A. Belaaouaj.
Neutrophil elastase contributes to cigarette smoke-induced emphysema in mice.
Am J Pathol, 163 (2003), pp. 2329-2335
[55]
H. Bostrom, K. Willetts, M. Pekny, P. Leveen, P. Lindahl, H. Hedstrand, et al.
PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis.
Cell, 85 (1996), pp. 863-873
[56]
M. Weinstein, X. Xu, K. Ohyama, C.X. Deng.
FGFR-3 and FGFR-4 function cooperatively to direct alveogenesis in the murine lung.
Development, 125 (1998), pp. 3615-3623
[57]
D.P. Wendel, D.G. Taylor, K.H. Albertine, M.T. Keating, D.Y. Li.
Impaired distal airway development in mice lacking elastin.
Am J Respir Cell Mol Biol, 23 (2000), pp. 320-326
[58]
D.G. Morris, X. Huang, N. Kaminski, Y. Wang, S.D. Shapiro, G. Dolganov, et al.
Loss of integrin alpha(v)beta6-mediated TGF-beta activation causes Mmp12-dependent emphysema.
Nature, 422 (2003), pp. 169-173
[59]
S.E. Wert, M. Yoshida, A.M. LeVine, M. Ikegami, T. Jones, G.F. Ross, et al.
Increased metalloproteinase activity, oxidant production, and emphysema in surfactant protein D gene-inactivated mice.
Proc Natl Acad Sci U S A, 97 (2000), pp. 5972-5977
[60]
K.J. Leco, P. Waterhouse, O.H. Sanchez, K.L. Gowing, A.R. Poole, A. Wakeham, et al.
Spontaneous air space enlargement in the lungs of mice lacking tissue inhibitor of metalloproteinases-3 (TIMP-3).
J Clin Invest, 108 (2001), pp. 817-829
[61]
D. Curiel, M. Brantly, E. Curiel, L. Stier, R.G. Crystal.
Alpha 1-antitrypsin deficiency caused by the alpha 1-antitrypsin Nullmattawa gene. An insertion mutation rendering the alpha 1-antitrypsin gene incapable of producing alpha 1-antitrypsin.
J Clin Invest, 83 (1989), pp. 1144-1152
[62]
A. Agusti, W. MacNee, K. Donaldson, M. Cosio.
Hypothesis: does COPD have an autoimmune component?.
Thorax, 58 (2003), pp. 832-834
[63]
M.G. Cosio, M. Saetta, A. Agusti.
Immunologic aspects of chronic obstructive pulmonary disease.
N Engl J Med, 360 (2009), pp. 2445-2454
[64]
B.G. Cosio, A. Agusti.
Autoimmunity in chronic obstructive pulmonary disease (COPD).
Arch Bronconeumol, 41 (2005), pp. 10-14
[65]
F. Kheradmand, M. Shan, C. Xu, D.B. Corry.
Autoimmunity in chronic obstructive pulmonary disease: clinical and experimental evidence.
Expert Rev Clin Immunol, 8 (2014), pp. 285-292
[66]
L. Taraseviciene-Stewart, R. Scerbavicius, K.H. Choe, M. Moore, A. Sullivan, M.R. Nicolls, et al.
An animal model of autoimmune emphysema.
Am J Respir Crit Care Med, 171 (2005), pp. 734-742
[67]
L. Taraseviciene-Stewart, N. Burns, D. Kraskauskas, M.R. Nicolls, R.M. Tuder, N.F. Voelkel.
Mechanisms of autoimmune emphysema.
Proc Am Thorac Soc, 3 (2006), pp. 486-487
[68]
L. Taraseviciene-Stewart, I.S. Douglas, P.S. Nana-Sinkam, J.D. Lee, R.M. Tuder, M.R. Nicolls, et al.
Is alveolar destruction and emphysema in chronic obstructive pulmonary disease an immune disease?.
Proc Am Thorac Soc, 3 (2006), pp. 687-690
[69]
S.H. Lee, S. Goswami, A. Grudo, L.Z. Song, V. Bandi, S. Goodnight-White, et al.
Antielastin autoimmunity in tobacco smoking-induced emphysema.
Nat Med, 13 (2007), pp. 567-569
[70]
Y.B. Kuo, C.A. Chang, Y.K. Wu, M.J. Hsieh, C.H. Tsai, K.T. Chen, et al.
Identification and clinical association of anti-cytokeratin 18 autoantibody in COPD.
Immunol Lett, 128 (2010), pp. 131-136
[71]
C.A. Feghali-Bostwick, A.S. Gadgil, L.E. Otterbein, J.M. Pilewski, M.W. Stoner, E. Csizmadia, et al.
Autoantibodies in patients with chronic obstructive pulmonary disease.
Am J Respir Crit Care Med, 177 (2008), pp. 156-163
[72]
C.A. Brandsma, W. Timens, M. Geerlings, H. Jekel, D.S. Postma, M.N. Hylkema, et al.
Induction of autoantibodies against lung matrix proteins and smoke-induced inflammation in mice.
BMC Pulm Med, 10 (2014), pp. 64
[73]
P.J. Barnes.
New anti-inflammatory targets for chronic obstructive pulmonary disease.
Nat Rev Drug Discov, 12 (2013), pp. 543-559
[74]
T. Stevens, K. Ekholm, M. Granse, M. Lindahl, V. Kozma, C. Jungar, et al.
AZD9668: pharmacological characterization of a novel oral inhibitor of neutrophil elastase.
J Pharmacol Exp Ther, 339 (2014), pp. 313-320
[75]
S. Medicherla, M.F. Fitzgerald, D. Spicer, P. Woodman, J.Y. Ma, A.M. Kapoun, et al.
p38alpha-selective mitogen-activated protein kinase inhibitor SD-282 reduces inflammation in a subchronic model of tobacco smoke-induced airway inflammation.
J Pharmacol Exp Ther, 324 (2008), pp. 921-929
[76]
C. Duong, H.J. Seow, S. Bozinovski, P.J. Crack, G.P. Anderson, R. Vlahos.
Glutathione peroxidase-1 protects against cigarette smoke-induced lung inflammation in mice.
Am J Physiol Lung Cell Mol Physiol, 299 (2014), pp. L425-L433
[77]
R. Vlahos, S. Bozinovski.
Glutathione peroxidase-1 as a novel therapeutic target for COPD.
Redox Rep, 18 (2014), pp. 142-149
[78]
T. Rangasamy, C.Y. Cho, R.K. Thimmulappa, L. Zhen, S.S. Srisuma, T.W. Kensler, et al.
Genetic ablation of Nrf2 enhances susceptibility to cigarette smoke-induced emphysema in mice.
J Clin Invest, 114 (2004), pp. 1248-1259
[79]
G.D. Massaro, D. Massaro.
Retinoic acid treatment partially rescues failed septation in rats and in mice.
Am J Physiol Lung Cell Mol Physiol, 278 (2000), pp. L955-L960
[80]
S.E. McGowan.
Contributions of retinoids to the generation and repair of the pulmonary alveolus.
Chest, 121 (2002), pp. 206S-208S
[81]
J.P. Muyal, V. Muyal, S. Kotnala, D. Kumar, H. Bhardwaj.
Therapeutic potential of growth factors in pulmonary emphysematous condition.
[82]
L. Martinez-Galan, L. del Puerto-Nevado, S. Perez-Rial, J.J. Diaz-Gil, N. Gonzalez-Mangado, G. Peces-Barba.
Liver growth factor improves pulmonary fibrosis secondary to cadmium administration in rats.
Arch Bronconeumol, 46 (2010), pp. 20-26
[83]
T.P. Cremona, S.A. Tschanz, C. von Garnier, C. Benarafa.
SerpinB1 deficiency is not associated with increased susceptibility to pulmonary emphysema in mice.
Am J Physiol Lung Cell Mol Physiol, 305 (2013), pp. L981-L989
[84]
P.R. Reynolds, M.G. Cosio, J.R. Hoidal.
Cigarette smoke-induced Egr-1 upregulates proinflammatory cytokines in pulmonary epithelial cells.
Am J Respir Cell Mol Biol, 35 (2006), pp. 314-319
[85]
T. Maeno, A.M. Houghton, P.A. Quintero, S. Grumelli, C.A. Owen, S.D. Shapiro.
CD8+ T Cells are required for inflammation and destruction in cigarette smoke-induced emphysema in mice.
J Immunol, 178 (2007), pp. 8090-8096
[86]
K. Aoshiba, N. Yokohori, A. Nagai.
Alveolar wall apoptosis causes lung destruction and emphysematous changes.
Am J Respir Cell Mol Biol, 28 (2003), pp. 555-562
[87]
J.L. Wright, H. Tai, A. Churg.
Vasoactive mediators and pulmonary hypertension after cigarette smoke exposure in the guinea pig.
J Appl Physiol, 100 (2006), pp. 672-678
[88]
Y. Takubo, A. Guerassimov, H. Ghezzo, A. Triantafillopoulos, J.H. Bates, J.R. Hoidal, et al.
Alpha1-antitrypsin determines the pattern of emphysema and function in tobacco smoke-exposed mice: parallels with human disease.
Am J Respir Crit Care Med, 166 (2002), pp. 1596-1603
[89]
A. Churg, R. Wang, X. Wang, P.O. Onnervik, K. Thim, J.L. Wright.
Effect of an MMP-9/MMP-12 inhibitor on smoke-induced emphysema and airway remodelling in guinea pigs.
Thorax, 62 (2007), pp. 706-713
[90]
G. John, K. Kohse, J. Orasche, A. Reda, J. Schnelle-Kreis, R. Zimmermann, et al.
The composition of cigarette smoke determines inflammatory cell recruitment to the lung in COPD mouse models.
Clin Sci, 126 (2014), pp. 207-221
[91]
C. Glynos, L.L. Dupont, T. Vassilakopoulos, A. Papapetropoulos, P. Brouckaert, A. Giannis, et al.
The role of soluble guanylyl cyclase in chronic obstructive pulmonary disease.
Am J Respir Crit Care Med, 188 (2013), pp. 789-799
[92]
S. Lea, J. Plumb, H. Metcalfe, D. Spicer, P. Woodman, J.C. Fox, et al.
The effect of peroxisome proliferator-activated receptor-gamma ligands on in vitro and in vivo models of COPD.
Eur Respir J, 43 (2014), pp. 409-420
[93]
K.R. Bracke, F.M. Verhamme, L.J. Seys, C. Bantsimba-Malanda, D.M. Cunoosamy, R. Herbst, et al.
Role of CXCL13 in cigarette smoke-induced lymphoid follicle formation and chronic obstructive pulmonary disease.
Am J Respir Crit Care Med, 188 (2013), pp. 343-355
[94]
P.L. Podolin, J.P. Foley, D.C. Carpenter, B.J. Bolognese, G.A. Logan, E. Long 3rd, et al.
T cell depletion protects against alveolar destruction due to chronic cigarette smoke exposure in mice.
Am J Physiol Lung Cell Mol Physiol, 304 (2013), pp. L312-L323
[95]
B.L. Eppert, B.W. Wortham, J.L. Flury, M.T. Borchers.
Functional characterization of T cell populations in a mouse model of chronic obstructive pulmonary disease.
J Immunol, 190 (2013), pp. 1331-1340

Please cite this article as: Pérez-Rial S, Girón-Martínez Á, Peces-Barba G. Modelos animales de enfermedad pulmonar obstructiva crónica. Arch Bronconeumol. 2015;51:121–127.

Copyright © 2014. SEPAR
Archivos de Bronconeumología
Article options
Tools

Are you a health professional able to prescribe or dispense drugs?