746
Views
131
CrossRef citations to date
0
Altmetric
Review

Animal models of asthma: utility and limitations

, , , &
Pages 293-301 | Published online: 07 Nov 2017

Abstract

Clinical studies in asthma are not able to clear up all aspects of disease pathophysiology. Animal models have been developed to better understand these mechanisms and to evaluate both safety and efficacy of therapies before starting clinical trials. Several species of animals have been used in experimental models of asthma, such as Drosophila, rats, guinea pigs, cats, dogs, pigs, primates and equines. However, the most common species studied in the last two decades is mice, particularly BALB/c. Animal models of asthma try to mimic the pathophysiology of human disease. They classically include two phases: sensitization and challenge. Sensitization is traditionally performed by intraperitoneal and subcutaneous routes, but intranasal instillation of allergens has been increasingly used because human asthma is induced by inhalation of allergens. Challenges with allergens are performed through aerosol, intranasal or intratracheal instillation. However, few studies have compared different routes of sensitization and challenge. The causative allergen is another important issue in developing a good animal model. Despite being more traditional and leading to intense inflammation, ovalbumin has been replaced by aeroallergens, such as house dust mites, to use the allergens that cause human disease. Finally, researchers should define outcomes to be evaluated, such as serum-specific antibodies, airway hyperresponsiveness, inflammation and remodeling. The present review analyzes the animal models of asthma, assessing differences between species, allergens and routes of allergen administration.

Introduction

Asthma affects approximately 300 million individuals of all age groups worldwide and its prevalence is increasing.Citation1 According to estimates, there are 18 asthma-related deaths per million people and 180,000 deaths per year.Citation2 Asthma has an impact on society because of adults’ loss of productivity and children’s learning impairment. Global Strategy for Asthma Management and Prevention defines “asthma as a heterogeneous disease, usually characterized by chronic airway inflammation. It is defined by the history of respiratory symptoms such as wheeze, shortness of breath, chest tightness and cough that vary over time and in intensity, together with variable expiratory airflow limitation”.Citation1 This chronic inflammation leads to airway remodeling, characterized by mucus hypersecretion, epithelial fibrosis, metaplasia and hyperplasia of goblet cells, and hypertrophy and hyperplasia of airway smooth muscle.Citation3

For many reasons, studies of humans with asthma do not fit all the ethical committee requirements. Therefore, animal models are necessary to better understand the pathophysiological mechanisms and to evaluate both safety and efficacy of new therapies on asthma before starting clinical trials in humans.Citation4 Nevertheless, the use of experimental animals in research laboratories also requires compliance with ethical precepts. These requirements were published by the National Institutes of Health (NIH), which were revised over 20 years ago (Guide for the Care and Use of Laboratory Animals – NIH; publication 85–23, revised in 1985). Since then, experimental models that do not meet these requirements are not acceptable.

Different phenotypes have been described in asthma but there is no standard way to distinguish them.Citation5 They differ regarding clinical parameters, physiological criteria and environmental triggers, and biomarkers to identify distinct endotypes are needed.Citation6 Animal models are limited for not being able to mimic all features and phenotypes of human asthma.Citation7,Citation8

However, they have proved their worth in amplifying the knowledge of many inflammatory, structural and physiological characteristics of asthma.Citation7 The Type 2 (T2 High) phenotype has been widely studied, but unfortunately in half of asthma patients the immune response is not Th2 mediated.Citation8,Citation9 Therefore, animal models that best represent each phenotype of asthma are needed.Citation7

There are many asthma models described in the literature, using different species and different methods to better mimic human asthma. They represent a scenario to understand disease pathophysiology and to test potential drug therapies. Positive results in animal experiments can be translated to clinical studies.Citation8,Citation10 In this review we perform a different approach about animal models of asthma, focusing on differences between species, allergens, routes of allergen administration and major outcomes evaluated.

Animal species

Asthma is a complex syndrome observed exclusively in humans. In animals, asthma-like conditions are observed in cats with eosinophilic bronchitis and in equines with heaves.Citation11,Citation12 During the last decades, several studies were performed using animal models to better understand the pathophysiology of disease and its immunological mechanisms.Citation13

Many animal species have been used to study the mechanisms involved in asthma (Drosophila, rat, guinea pig, cat, dog, swine, cattle, sheep, horse and primates), but the most common model is the murine allergic airway inflammation.Citation14Citation16

Fruit fly Drosophila melanogaster has being used as an alternative prototype to address the innate immunity and airway epithelial cells in asthma.Citation17 This invertebrate model merges a comparatively simple physiology and genetic organization together with an unparalleled toolkit for genetic manipulation. Roeder et al proposed the use of Drosophila as an asthma model based on the potential homology of known asthma-susceptibility genes between humans and this invertebrate as well as the characteristics of airway immunity and asthma-like phenotypes observed in the fruit fly.Citation18

Experimental asthma in guinea pigs was introduced in 1937 by Kallós P and Kallós L.Citation19 Guinea pigs have been the animal species most widely used as the first animal models of allergic respiratory disease, because they present airway physiological processes much similar to humans. They also respond strongly to allergens and have autonomic control of airways. Then, efficacy of drugs such as bronchodilators can be tested before their use in clinical trials.Citation18

On the other hand, primary disadvantages of using guinea pig are the lack of specific probes and reagents for studying allergic outcomes, which are not easily available. So, comprehension of humoral and cellular mechanisms was very difficult. In addition, there is paucity of transgenic models and few strains of guinea pigs for comparative studies.Citation20 Moreover, the notable axon reflex presented in these animals has not been described in human airways until now.Citation20 Another handicap of guinea pig is the longer gestation time (60–75 days) comparing with mice (20–30 days), and the lower number of offsprings.

In 1994, the first mouse models resembling allergic asthma were published and, thereafter, have resulted in significant strides in our understanding of atopic disease pathophysiology.Citation21Citation25 Mice have become the most widely used species, because they are easy to breed, maintain and handle. In addition, a wide array of specific reagents are available for analysis of cellular and humoral responses, and genetically engineered transgenic or gene-knockout mice for modeling airway disease are available.Citation26,Citation27 The most commonly used mouse strain in antigen challenge models is BALB/c as they develop a good Th2-biased immunological response, although C57BL/6 and A/J strains have also been used successfully in experimental models of respiratory allergic disease.Citation28

Mice do not develop asthma spontaneously. So, the disease has to be artificially induced in the airways. The murine models of allergic respiratory diseases induced by ovalbumin (OVA) and aeroallergens have been widely used to elucidate immunological and nonimmunological mechanisms involved in the pathogenesis of asthma. In addition, they are useful for identifying and investigating new targets for controlling allergic inflammation.Citation28 For that, acute and chronic experimental models have been developed.

Acute mice models have successfully reproduced many features of asthma such as high levels of serum total and specific immunoglobulin E (IgE), airway inflammation, epithelial hypertrophy, goblet cell hyperplasia and airway hyper-responsiveness (AHR). Besides, in some models, researches can induce early- and late-phase bronchoconstriction in response to allergen challenge. On the other hand, in acute models, the pattern and distribution of pulmonary inflammation are different from that found in asthmatic individuals.Citation29 First, bronchoalveolar lavage (BAL) and histologic studies indicate that the influx of inflammatory cells is dominated by eosinophils. Many pathologic findings of chronic human asthma, such as chronic inflammation of the airway wall and remodeling, cannot be observed since the animal is exposed to the antigen-less times in acute models. Finally, there is a major difference between acute mouse models and human asthma: airway inflammation and AHR seem to resolve within a few weeks after the final antigen provocation in the animal. In human asthma, inflammation persists and a new exposition to the allergen tends to induce recurrence of symptoms.Citation28

Although presenting some discrepancies, the acute asthma models were successfully used to investigate some of the pathophysiological aspects of this disease. For example, cell-mediated pulmonary inflammation and many disease mediators have been well demonstrated. One of the major theories proved by acute animal models is that allergic asthma is a Th2-mediated disease. Moreover, these studies allow the comprehension about the role of the T cell and eosinophils in the occurrence of allergic response and AHR.Citation28

Chronic models of murine asthma have been shown to reproduce more closely human asthma. Some of the hallmarks are allergen-dependent sensitization, a Th2-dependent allergic inflammation characterized by eosinophilic influx into the airway mucosa and AHR. Thus, chronic models of asthma lead to remodeling of the airways, which is one of the observations of asthma in human adults. In addition, in some models of chronic asthma, AHR and pulmonary inflammation persist for days or weeks after the last allergen challenge.Citation4,Citation29,Citation30 However, maintenance of AHR and lung inflammation vary according to the exposure protocol applied.Citation28,Citation31

As noted above, asthma is a peculiar human disease and no laboratory animal commonly used to study this condition, including mice, rats, guinea pigs or rabbits, exhibits similar symptoms of human asthma. The main exception is eosinophilic bronchitis presented by cats.Citation11

In terms of T-helper activation profile, according to cytokine measurement, few data are available about most species. In rodent models, Th2 cytokine profile (interleukin 4 [IL-4], IL-5 and IL-13) has been well documented.Citation15,Citation20,Citation32 In a feline model, IL-4 has been demonstrated in airway allergic inflammation,Citation33 while in a sheep asthma model recently published, it has been demonstrated that both IL-4 and IL-13 increase in BAL after allergen challenge.Citation34 In equines, former studies were controversial in terms of inflammation pattern, ranging from Th1, Th2 and Th17 cytokines profile,Citation35,Citation36 but recent studies have shown that inflammation seems to be mainly mediated by Th17 cytokines, like CXCL13 and IL-17.Citation37,Citation38

In , we summarize some of the main discrepancies between different animal species used in animal models.

Table 1 Advantages and disadvantages of animal species most frequently found in experimental models of asthma

Allergens and agents

Studies with animal models of allergic asthma assess disease pathogenesis. It is well known that allergic imune response initiates with a first phase named sensitization, which is characterized by production of specific IgE driven to the allergen by B cells. Once IgE is produced, it will bind to the high-affinity receptor FcεR1 on the surface of mast cells and basophils.Citation32,Citation33

The second phase is named challenge. In future contacts with the same allergen, effector cells (mast cells and basophils) in the airways will be activated through FcεR1, initiating an immediate hypersensitivity reaction.Citation40,Citation41 Minutes after allergen cross-linking two IgE molecules, these effector cells release preformed and rapidly synthesized mediators such as histamine, resulting in bronchospasm, edema and mucous secretion in the lower airways.Citation41 There can be a late phase, which is mediated by cytokines and chemokines and is characterized by edema and leukocytic influx, usually 6–24 hours after the immediate phase. The most important leukocytes of the late phase are eosinophils, which are recruited by IL-5 and are essential to maintain the chronic inflammatory process and tissue damage. As asthma is a chronic disease, recurrence of challenges leads to chronic eosinophilic inflammation.Citation41

To mimic the pathogenesis of human asthma, protocols for development of animal models must include a sensitization and a challenge phase.Citation42 They usually use repeated doses of systemic allergen administration together with adjuvants, such as aluminum hydroxide, to increase immune response.Citation28 However, as cited above, the pattern of lung inflammation and its distribution within lower airways is quite different from human asthma.Citation43 There are many sensitization protocols that can induce acute or chronic asthma in animals and they will be addressed later on.

Adjuvants, such as potassium aluminum sulfate, are used to increase allergen immunogenicity leading to better chances of sensitization.Citation44 In human beings, sensitization relies on mucosa exposure to allergen followed by immunological recognition and Th2 inflammatory response. In animal models, allergens can be delivered to the immune system through subcutaneous (SC) injection, intraperitoneal (IP) injection, intranasal (IN) drops or inhaling.

Allergens that have been used in animal models are OVA, house dust mite (HDM), such as Dermatophagoides pteronyssinus (Der p) or D. farinae (Der f), mite allergens (Der p 1, Der f 1, Der p 23, etc), fungi (Aspergillus fumigatus, Alternaria alternata), cockroach extracts, Ascaris antigens, cotton dust, ragweed and latex (Hevea brasiliensis). The allergen of choice depends on the condition to be replicated and it can be used separately or in combination.Citation15

OVA is the most used allergen. It is derived from chicken egg and can be produced in large quantities, making it less expensive. OVA has been used in experimental models of asthma and induces intense allergic pulmonary inflammation.Citation45 Nevertheless, OVA does not induce airway inflammation in humans and it has been questioned as a good allergen to study asthma. HDM has been successfully used to induce asthma in animal models. Three characteristics of this allergen make it suitable: intrinsic enzymatic activity, immunogenicity and direct activation through the Dectin-2 receptor of innate immune cells that promote allergic inflammation.Citation46,Citation47

Cockroach extracts mostly used in animal models are derived from Blatella germanica. Previous studies have shown that the protein Bla g 2 is a potent allergen and it can be identified in 60%–80% of patients allergic to domestic cockroaches.Citation48

Ascaris lumbricoides is one of the most common parasites found in human disease, infecting about 25% of the world’s population. Ascaris allergens were described over two decades ago.Citation49 Since then, their antigens have been used in a few animal models of asthma.Citation50Citation53

It is well known that fungi are between the major allergens that induce allergic rhinitis and asthma. Then, many animal models have been developed, particularly using A. fumigatus, the classical causative agent of allergic bronchopulmonary aspergillosiss.Citation54 Animals that underwent Aspergillus sensitization and challenge develop high levels of IgE, eosinophilia and lung inflammation.Citation54

Ragweeds are flowering plants from the genus Ambrosia, belonging to the aster family Asteraceae. Ragweed pollen is responsible for allergic reactions in humans, particularly hay fever.Citation55 It is estimated that half of the cases of hay fever in North America are induced by ragweed.Citation55 Despite the impact of pollen allergy, few ragweed models can be found in the literature. One of them is a dog model with T cells locally activated in the lungs within 4 hours after exposure to ragweed allergen.Citation56

Proteins from H. brasiliensis (latex) can also lead to sensitization and allergic reactions in human.Citation57 Results obtained from a murine asthma model induced by latex suggest that curcumin has potential therapeutic effects. The study showed that eosinophilic inflammation, expression of co-stimulatory molecules and expression of some genes involved in the process were attenuated by curcumin.Citation58

Routes of sensitization and challenge

Chronic models of allergic respiratory disease involve repeated airway exposition to low levels of allergen for periods of up to 12 weeks. Different antigens have been employed and coadministration of an adjuvant is usually, but not always, required.Citation28 On the other hand, repeated long-term allergen exposure, in particular with protein antigens such as OVA, may be associated with tolerance development.Citation59

Aiming to develop both acute and chronic animal models of asthma, several approaches in terms of routes of sensitization and challenge have been tested. Since 1980s, IP route has probably been the most traditional way to induce sensitization. One of the most commonly repeated protocols was animal sensitization with two IP injections spaced by 7–14 days and the challenge is performed 1 week later with the culprit allergen.Citation51Citation54

Nevertheless, SC route has been successfully used in last years, both in models of OVA and aeroallergen-induced pulmonary inflammation. As far as we know, the first manuscript about an animal model of asthma using SC injections for sensitization was published in 1999.Citation61 Since then, many studies were performed, but there is limited data in terms of comparison between these two different systemic routes, IP and SC.

SC and IP routes have been compared in terms of sensitization with OVA, with conflicting results,Citation62,Citation63 but until recently there were no published data about this comparison in animal models of asthma induced by aeroallergens. In 2015, we showed that sensitization by SC route was superior than IP in a murine model of asthma induced by HDM.Citation64 Nevertheless, to our knowledge, it is still the only publication on this topic, and further studies comparing different allergens and protocols are needed to confirm our findings.

After systemic sensitization, allergen challenge is necessary to drive inflammation to the airways. Most studies published to date use allergen challenge via the airways, usually over a period of several days. Allergen may be inhaled as a nebulized formulation (aerosol), or administered by intratracheal (IT) or IN instillation of an aqueous formulation.Citation50,Citation59Citation61 In authors’ experimental practice, aerosol route of challenge spends higher amount of allergen, but is less invasive and does not require animal sedation. On the other hand, IN and IT routes are more invasive and require sedation to be administered. The clear advantage of IN and IT routes is that allergens are instillated directly inside the airways and could drive a more intense allergic inflammation.

One of the major criticisms over animal models of asthma is that they do not mimic the real ways to induce the allergic response. First of all, it is well known that asthma is a chronic disease resulting from intermittent or continued aeroallergen exposure leading to airway inflammation. This exposure occurs throughout life, primarily via the inhalation of allergens and irritants through the airways during ventilation. Furthermore, using IP or SC routes to sensitize animals is far from natural in terms of inducing an allergic inflammatory response. In line with that knowledge, some studies have evaluated the possibility to use IN instillations of allergen to sensitize animals and then challenge with aerosol.Citation68,Citation69 IN instillation of allergen would be the most similar to that occurring in human asthma. Two different protocols published in 2004 used repeated IN exposition to HDM without adjuvants and induce pulmonary allergic inflammation analog to asthma.Citation30,Citation70 The study published by Johnson et alCitation30 used a chronic protocol, with IN instillation of HDM or OVA 5 days a week, during seven consecutive weeks. They showed that HDM, but not OVA, elicited severe and persistent eosinophilic airway inflammation, suggesting that continuous exposure to OVA could have led to tolerance or the need of an adjuvant for sensitization.Citation30 Chronic exposure to aeroallergen mimics better human asthma, and could allow the development of better treatment and immunotherapeutic strategies. Those reasons explain why this protocol has recently been replicated or adapted so many times in the literature.Citation71Citation76

In terms of sensitization and challenge, we can conclude that using the airways to administer the allergen has been a recent tendency, trying to mimic human asthma, instead of IP or SC routes. However, SC route can be very interesting to study new immunotherapeutic strategies, taking into consideration that SC is the most widely used route of allergen immunotherapy in humans. In , we describe the main differences between routes of sensitization and challenge.

Table 2 Comparison between different routes of sensitization and challenge in animal models of asthma

Major outcomes

In general, animal models have contributed to the current understanding of how the immune system interacts with the functional respiratory system and pulmonary pathophysiology. Differences in observed results may be related to distinct allergens, sensitization routes, experimental designs and animal species or lineages used. Anyway, most of the experimental asthma models usually evaluate the following outcomes: immunological (IgE, IgG, cytokines), histopathological (pattern of inflammatory infiltrate in the airway) and functional (lung function measured by plethysmography).

Several authors have shown that allergen-induced respiratory disease alters lung function with changes in airway resistance, airway elastance, or increased hyperresponsiveness.Citation77Citation79 This alteration in lung function is followed by an increased deposition of elastic and collagen fibers in the perivascular space and in parenchyma lung tissue,Citation80 goblet cell hyperplasia and airway smooth muscle thickening similar to the pathologies observed in human asthma.Citation75Citation77

Regarding pulmonary inflammation, it is possible to observe in experimental models of asthma an intense influx of eosinophils, especially in the airways, peribronchial space and parenchyma.Citation58,Citation81Citation85 Increased eosinophil count is also present in blood and BAL in conjunction with an increase of lymphocytes, macrophages and neutrophils.Citation86Citation88 Concomitantly, dendritic cells may migrate from outside into the ganglia to interact with sensory neurons enhancing or protecting the allergic airway inflammation.Citation89

Cytokine production is another important outcome assessed in experimental models of asthma. Influx of eosinophil and other leukocytes, as well as the role of many cytokines and chemokines has been demonstrated through animal models.Citation90,Citation91 In recent years, biological actions of some novel mediators, such as interleukins IL-25 and IL-33 and thymic stromal lymphopoietin, major airway epithelial-derived cytokines, have been described. These cytokines have been entitled as “epithelial-derived alarmins” because of the ability of activation and potentiation of the immune system. An intense correlation of these epithelial-derived alarmins with the pathobiological responses induced by aeroallergens was observed in the airways.Citation92 Finally, about humoral response, guinea pigs share with mice the shortcoming of utilizing IgG1 and IgE in regulating the immediate hypersensitivity response to allergen.Citation11Citation14,Citation19,Citation20,Citation93Citation98

Conclusion

Animal models remain the easiest way to understand pathophysiology of allergic asthma and to help developing new drugs and immunotherapy strategies for the treatment of this complex disease. However, there are tremendous variations between animal species, protocols and allergens used, but few studies assessed these discrepancies in order to determine the best model. The most recent protocols have been inducing sensitization and challenge by the same routes in which the human disease occurs (eg, IN route) and to using the same aeroallergens that trigger clinical disease (eg, HDMs). Nevertheless, many approaches cannot be simply translated to human disease. Researchers should take a step back to define what the best strategies are. Then they will be ready to move forward, to better develop models of different asthma phenotypes, such as non-atopic disease, and to advance further in the development of future treatments. It seems that transgenic models will be great future options for a better understanding of the role of each molecule and cytokine in the different asthma phenotypes and endotypes.

Disclosure

The authors report no conflicts of interest in this work.

References

  • Global Initiative for AsthmaGlobal Strategy for Asthma Management and Prevention (updated 2016)GINA2016 Available from: http://ginasthma.org/wp-content/uploads/2016/04/wms-GINA-2016-main-report-final.pdfAccessed October 5, 2017
  • WHOWHO Fact Sheet Bronchial AsthmaGenevaWorld Health Organization2000 No 206
  • ShinagawaKKojimaMMouse model of airway remodeling: strain differencesAm J Respir Crit Care Med2003168895996712857720
  • KumarRKHerbertCYangMKoskinenAMMcKenzieANFosterPSRole of interleukin-13 in eosinophil accumulation and airway remodelling in a mouse model of chronic asthmaClin Exp Allergy20023271104111112100061
  • WenzelSEAsthma: defining of the persistent adult phenotypesLancet2006368953780481316935691
  • HesselmarBEnelundACErikssonBPadyukovLHansonLÅAbergNThe heterogeneity of asthma phenotypes in children and young adultsJ Allergy (Cairo)2012201216308922577403
  • ChapmanDGTullyJENolinJDJanssen-HeiningerYMIrvinCGAnimal models of allergic airways disease: where are we and where to next?J Cell Biochem2014115122055206425043224
  • SagarSAkbarshahiHUllerLTranslational value of animal models of asthma: Challenges and promisesEur J Pharmacol201575927227725823808
  • MullaneKWilliamsMAnimal models of asthma: reprise or reboot?Biochem Pharmacol201487113113923831953
  • SzelenyiIAnimal models of bronchial asthmaInflamm Res2000491263965411211914
  • HerszbergBRamos-BarbónDTamaokaMMartinJGLavoieJPHeaves, an asthma-like equine disease, involves airway smooth muscle remodelingJ Allergy Clin Immunol2006118238238816890762
  • WilliamsKRomanJStudying human respiratory disease in animals role of induced and naturally occurring modelsJ Pathol2016238222023226467890
  • van der WorpHBHowellsDWSenaESCan animal models of disease reliably inform human studies?PLoS Med201073e100024520361020
  • BlumeCDaviesDEIn vitro and ex vivo models of human asthmaEur J Pharm Biopharm201384239440023313714
  • ZoskyGRSlyPDAnimal models of asthmaClin Exp Allergy200737797398817581191
  • KirschvinkNReinholdPUse of alternative animals as asthma modelsCurr Drug Targets20089647048418537586
  • LemaitreBHoffmannJThe host defense of Drosophila melanogasterAnnu Rev Immunol20072569774317201680
  • RoederTIsermannKKabeschMDrosophila in asthma researchAm J Respir Crit Care Med20091791197998319342413
  • KallósPKallósLExperimental asthma in guinea pigs revisitedInt Arch Allergy Appl Immunol198473177856141145
  • CanningBJChouYUsing guinea pigs in studies relevant to asthma and COPDPulm Pharmacol Ther200821570272018462968
  • EpsteinMMDo mouse models of allergic asthma mimic clinical disease?Int Arch Allergy Immunol200413318410014726635
  • KungTTJonesHAdamsGK3rdCharacterization of a murine model of allergic pulmonary inflammationInt Arch Allergy Immunol1994105183908086833
  • BrusselleGGKipsJCTavernierJHAttenuation of allergic airway inflammation in IL-4 deficient miceClin Exp Allergy199424173808156448
  • GavettSHChenXFinkelmanFWills-KarpMDepletion of murine CD4+ T lymphocytes prevents antigen-induced airway hyper-reactivity and pulmonary eosinophiliaAm J Respir Cell Mol Biol19941065875938003337
  • LukacsNWStrieterRMChensueSWKunkelSLInterleukin-4-dependent pulmonary eosinophil infiltration in a murine model of asthmaAm J Respir Cell Mol Biol19941055265328179915
  • ShapiroSDThe use of transgenic mice for modeling airways diseasePulm Pharmacol Ther200821569970118407535
  • Bonamichi-SantosRAunMVAgondiRCKalilJGiavina-BianchiPMicrobiome and asthma: what have experimental models already taught us?, microbiome and asthma: what have experimental models already taught us?J Immunol Res20152015e614758
  • NialsATUddinSMouse models of allergic asthma: acute and chronic allergen challengeDis Model Mech200814–521322019093027
  • McMillanSJLloydCMProlonged allergen challenge in mice leads to persistent airway remodellingClin Exp Allergy200434349750715005746
  • JohnsonJRWileyREFattouhRContinuous exposure to house dust mite elicits chronic airway inflammation and structural remodelingAm J Respir Crit Care Med2004169337838514597485
  • LloydCMBuilding better mouse models of asthmaCurr Allergy Asthma Rep20077323123617448336
  • HanRTKimSChoiKAsthma-like airway inflammation and responses in a rat model of atopic dermatitis induced by neonatal capsaicin treatmentJ Asthma Allergy20171018118928572736
  • DeClueAESchooleyENafeLAReineroCRfeG-COOH blunts eosinophilic airway inflammation in a feline model of allergic asthmaInflamm Res200958845746219255722
  • LiraviBPiedrafitaDNguyenGBischofRJDynamics of IL-4 and IL-13 expression in the airways of sheep following allergen challengeBMC Pulm Med20151510126362930
  • Lavoie-LamoureuxAMoranKBeauchampGIL-4 activates equine neutrophils and induces a mixed inflammatory cytokine expression profile with enhanced neutrophil chemotactic mediator release ex vivoAm J Physiol Lung Cell Mol Physiol20102994L472L48220639353
  • RiihimäkiMRaineAArtTLekeuxPCouëtilLPringleJPartial divergence of cytokine mRNA expression in bronchial tissues compared to bronchoalveolar lavage cells in horses with recurrent airway obstructionVet Immunol Immunopathol20081223–425626418243337
  • PacholewskaAJagannathanVDrögemüllerMImpaired cell cycle regulation in a natural equine model of asthmaPLoS One2015108e013610326292153
  • KornAMillerDDongLBucklesELWagnerBAinsworthDMDifferential gene expression profiles and selected cytokine protein analysis of mediastinal lymph nodes of horses with chronic recurrent airway obstruction (RAO) support an interleukin-17 immune responsePLoS One20151011e014262226561853
  • KumarRKFosterPSST2: marker, activator and regulator of Th2 immunity?Clin Exp Allergy200232101394139612372115
  • Giavina-BianchiPAunMVTakejimaPKalilJAgondiRCUnited airway disease: current perspectivesJ Asthma Allergy201699310027257389
  • StoneKDPrussinCMetcalfeDDIgE, mast cells, basophils, and eosinophilsJ Allergy Clin Immunol20101252 Suppl 2S73S8020176269
  • PatelKNChorawalaMRAnimal models of asthmaJ Pharm Res Opin201415139147
  • KumarRKFosterPSModeling allergic asthma in mice: pitfalls and opportunitiesAm J Respir Cell Mol Biol200227326727212204888
  • IshidaWFukudaKSumiTAdjuvants determine the contribution of basophils to antigen sensitization in vivoImmunol Lett20111361495421147167
  • FuchsBBraunAImproved mouse models of allergy and allergic asthma chances beyond ovalbuminCurr Drug Targets20089649550218537588
  • KimCHAhnJHKimSJCo-administration of vaccination with DNA encoding T cell epitope on the Der p and BCG inhibited airway remodeling in a murine model of chronic asthmaJ Asthma200643534535316801138
  • BarrettNAMaekawaARahmanOMAustenKFKanaokaYDectin-2 recognition of house dust mite triggers cysteinyl leukotriene generation by dendritic cellsJ Immunol200918221119112819124755
  • ArrudaLKVailesLDMannBJMolecular cloning of a major cockroach (Blattella germanica) allergen, Bla g 2. Sequence homology to the aspartic proteasesJ Biol Chem19952703319563195687642642
  • O’DonnellIJMitchellGFAn investigation of the allergens of Ascaris lumbricoides using a radioallergosorbent test (RAST) and sera of naturally infected humans: comparison with an allergen for mice identified by a passive cutaneous anaphylaxis testAust J Biol Sci1978315459487751627
  • OhruiTSekizawaKAikawaTYamauchiKSasakiHTakishimaTVascular permeability and airway narrowing during late asthmatic response in dogs treated with MetopironeJ Allergy Clin Immunol19928959339431583251
  • FornhemCKumlinMLundbergJMAlvingKAllergen-induced late-phase airways obstruction in the pig: mediator release and eosinophil recruitmentEur Respir J199587110011097589393
  • KirschvinkNLeemansJDelvauxFSnapsFClercxCGustinPFunctional, inflammatory and morphological characterisation of a cat model of allergic airway inflammationVet J2007174354155317306576
  • MisawaMTakenouchiKAbiruTYoshinoYYanauraSStrain difference in an allergic asthma model in ratsJpn J Pharmacol198745163683119903
  • TakazonoTSheppardDCAspergillus in chronic lung disease: modeling what goes on in the airwaysMed Mycol2017551394727838644
  • TaramarcazPLambeletBClotBKeimerCHauserCRagweed (Ambrosia) progression and its health risks: will Switzerland resist this invasion?Swiss Med Wkly200513537–3853854816333764
  • OutTAWangSZRudolphKBiceDELocal T-cell activation after segmental allergen challenge in the lungs of allergic dogsImmunology2002105449950811985670
  • VandenplasORaulfMOccupational latex allergy: the current state of affairsCurr Allergy Asthma Rep20171731428251426
  • KurupVPBarriosCSRajuRJohnsonBDLevyMBFinkJNImmune response modulation by curcumin in a latex allergy modelClin Mol Allergy20075117254346
  • KumarRKHerbertCFosterPSThe “classical” ovalbumin challenge model of asthma in miceCurr Drug Targets20089648549418537587
  • WagersSLundbladLKEkmanMIrvinCGBatesJHThe allergic mouse model of asthma: normal smooth muscle in an abnormal lung?J Appl Physiol (1985)20049662019202714660507
  • CarvalhoCJancarSMarianoMSiroisPA rat model presenting eosinophilia in the airways, lung eosinophil activation, and pulmonary hyperreactivityExp Lung Res199925430331610378102
  • ConradMLYildirimAOSonarSSComparison of adjuvant and adjuvant-free murine experimental asthma modelsClin Exp Allergy20093981246125419438585
  • RodriguesAMSchmidtCZGualdiLPProposed short-term model of acute allergic response, without adjuvant use, in the lungs of miceJ Bras Pneumol2012385595604 Portuguese23147052
  • AunMVSaraiva-RomanholoBMAlmeidaFMSensitization by subcutaneous route is superior to intraperitoneal route in induction of asthma by house dust mite in a murine modeEinstein (Sao Paulo)201513456056626761554
  • StevensonCSBirrellMAMoving towards a new generation of animal models for asthma and COPD with improved clinical relevancePharmacol Ther201113029310521074553
  • LiaoECHoCMYinSCTsaiJJImmune responses to tyrophagus putrescentiae-induced airway inflammation in miceJ Investig Allergol Clin Immunol20132312029
  • Asai-TajiriYMatsumotoKFukuyamaSSmall interfering RNA against CD86 during allergen challenge blocks experimental allergic asthmaRespir Res20141513225344652
  • KriegerSMBoverhofDRWoolhiserMRHotchkissJAAssessment of the respiratory sensitization potential of proteins using an enhanced mouse intranasal test (MINT)Food Chem Toxicol20135916517623747714
  • PetersMKöhler-BachmannSLenz-HabijanTBufeAInfluence of an Allergen-specific Th17 response on remodeling of the airwaysAm J Respir Cell Mol Biol201654335035826222011
  • CatesECFattouhRWattieJIntranasal exposure of mice to house dust mite elicits allergic airway inflammation via a GM-CSF-mediated mechanismJ Immunol2004173106384639215528378
  • Piñeiro-HermidaSGregoryJALópezIPAttenuated airway hyperresponsiveness and mucus secretion in HDM-exposed Igf1r-deficient miceAllergy20177291317132628207927
  • SchmitDLeDDHeckSAllergic airway inflammation induces migration of mast cell populations into the mouse airwayCell Tissue Res Epub2017325
  • ZaffiniRDi PaolaRCuzzocreaSMenegazziMPARP inhibition treatment in a nonconventional experimental mouse model of chronic asthmaNaunyn Schmiedebergs Arch Pharmacol2016389121301131327604227
  • ShimaKKoyaTTsukiokaKEffects of sublingual immunotherapy in a murine asthma model sensitized by intranasal administration of house dust mite extractsAllergol Int2017661899627397923
  • HoffmanSMQianXNolinJDAblation of glutaredoxin-1 modulates house dust mite-induced allergic airways disease in miceAm J Respir Cell Mol Biol201655337738627035878
  • HoffmanSMNolinJDJonesJTAblation of the thiol transferase glutaredoxin-1 augments protein S-glutathionylation and modulates type 2 inflammatory responses and IL-17 in a house dust mite model of allergic airway disease in miceAnn Am Thorac Soc201613Suppl 1S97
  • NesiRTKennedy-FeitosaELanzettiMInflammatory and oxidative stress markers in experimental allergic asthmaInflammation20174041166117628391514
  • LundbladLKAGülecNPoynterMEThe role of iNKT cells on the phenotypes of allergic airways in a mouse modelPulm Pharmacol Ther201745808928483562
  • Alberca-CustódioRWGreiffoFRMacKenzieBAerobic exercise reduces asthma phenotype by modulation of the leukotriene pathwayFront Immunol2016723727379098
  • ToledoACSakodaCPPeriniAFlavonone treatment reverses airway inflammation and remodelling in an asthma murine modelBr J Pharmacol201316871736174923170811
  • ToledoACArantes-CostaFMMacchioneMSalbutamol improves markers of epithelial function in mice with chronic allergic pulmonary inflammationRespir Physiol Neurobiol2011177215516121443970
  • WaghADSharmaMMahapatraJChatterjeeAJainMAddepalliVInvestigation into the role of PI3K and JAK3 kinase inhibitors in murine models of asthmaFront Pharmacol201788228293189
  • BoskabadyMHAdel-KardanSIncreased muscarinic receptor blockade by atropine in tracheal chains of ovalbumin-sensitized guinea pigsPharmacology199958630030810325575
  • BuelsKSJacobyDBFryerADNon-bronchodilating mechanisms of tiotropium prevent airway hyperreactivity in a guinea-pig model of allergic asthmaBr J Pharmacol201216551501151421871018
  • BlonderJPMutkaSCSunXPharmacologic inhibition of S-nitrosoglutathione reductase protects against experimental asthma in BALB/c mice through attenuation of both bronchoconstriction and inflammationBMC Pulm Med201414324405692
  • UlrichKHincksJSWalshRAnti-inflammatory modulation of chronic airway inflammation in the murine house dust mite modelPulm Pharmacol Ther200821463764718407534
  • VenkayyaRLamMWillkomMGrünigGCorryDBErleDJThe Th2 lymphocyte products IL-4 and IL-13 rapidly induce airway hyper-responsiveness through direct effects on resident airway cellsAm J Respir Cell Mol Biol200226220220811804871
  • LewisCAJohnsonABroadleyKJEarly and late phase broncho-constrictions in conscious sensitized guinea-pigs after macro- and microshock inhalation of allergen and associated airway accumulation of leukocytesInt J Immunopharmacol1996186–74154229024944
  • LeDDRochlitzerSFischerAAllergic airway inflammation induces the migration of dendritic cells into airway sensory gangliaRespir Res2014157324980659
  • AkdisMAabAAltunbulakliCInterleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: receptors, functions, and roles in diseasesJ Allergy Clin Immunol20161384984101027577879
  • GourNWills-KarpMIL-4 and IL-13 signaling in allergic airway diseaseCytokine2015751687826070934
  • MitchellPDO’ByrnePMEPithelial derived cytokines in asthmaChest201715161338134427818325
  • OettgenHCMartinTRWynshaw-BorisADengCDrazenJMLederPActive anaphylaxis in IgE-deficient miceNature199437064883673708047141
  • MiyajimaIDombrowiczDMartinTRRavetchJVKinetJPGalliSJSystemic anaphylaxis in the mouse can be mediated largely through IgG1 and Fc gammaRIII. Assessment of the cardiopulmonary changes, mast cell degranulation, and death associated with active or IgE- or IgG1-dependent passive anaphylaxisJ Clin Invest19979959019149062348
  • PeriniAMotaIThe production of IgE and IgG1 antibodies in guinea-pigs immunized with antigen and bacterial lipopolysaccharidesImmunology19732522973054354826
  • OvaryZCaiazzaSSKojimaSPCA reactions with mouse antibodies in mice and ratsInt Arch Allergy Appl Immunol197548116211116878
  • HamelmannETakedaKSchwarzeJVellaATIrvinCGGelfandEWDevelopment of eosinophilic airway inflammation and airway hyper-responsiveness requires interleu- kin-5 but not immunoglobulin E or B lymphocytesAm J Respir Cell Mol Biol19992148048910502558
  • RegalJFImmunoglobulin G- and immunoglobulin E-mediated airway smooth muscle contraction in the guinea pigJ Pharmacol Exp Ther198422811161206694096