Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3090 2022-04-07 18:09:23 |
2 format correct -2 word(s) 3088 2022-04-08 02:44:36 | |
3 format correct Meta information modification 3088 2022-04-08 02:45:48 | |
4 format correct Meta information modification 3088 2022-04-08 02:46:42 | |
5 format correct Meta information modification 3088 2022-04-08 02:47:12 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Simões, J.; , .; Tilley, P. The Immune Mechanisms of Severe Equine Asthma. Encyclopedia. Available online: https://encyclopedia.pub/entry/21472 (accessed on 08 September 2024).
Simões J,  , Tilley P. The Immune Mechanisms of Severe Equine Asthma. Encyclopedia. Available at: https://encyclopedia.pub/entry/21472. Accessed September 08, 2024.
Simões, Joana, , Paula Tilley. "The Immune Mechanisms of Severe Equine Asthma" Encyclopedia, https://encyclopedia.pub/entry/21472 (accessed September 08, 2024).
Simões, J., , ., & Tilley, P. (2022, April 07). The Immune Mechanisms of Severe Equine Asthma. In Encyclopedia. https://encyclopedia.pub/entry/21472
Simões, Joana, et al. "The Immune Mechanisms of Severe Equine Asthma." Encyclopedia. Web. 07 April, 2022.
The Immune Mechanisms of Severe Equine Asthma
Edit

Severe equine asthma is a chronic respiratory disease of adult horses, occurring when genetically susceptible individuals are exposed to environmental respirable particles. This results in airway inflammation, mucus accumulation and bronchial constriction. Affected horses present with cough, nasal discharge and increased respiratory effort at rest. Although a complex diversity of genetic and immunological pathways contribute to the disease, these remain to be fully understood. 

severe equine asthma immunology

1. Introduction

Severe equine asthma (sEA) is a naturally occurring chronic respiratory disease [1], affecting up to 20% of adult horses in the Northern hemisphere [2]. Disease develops upon exposure of genetically susceptible individuals to environments with high concentrations of airborne respirable particles, capable of inducing airway inflammation [3]. A vast array of antigens have been implicated in the etiology of sEA and it is thought that airway inflammation results from the synergistic effect of multiple allergens [4], to which individuals are susceptible in unique ways. This disease has been mostly associated with hay feeding and stabling, being termed as stable-associated sEA, but summer pasture-associated sEA also occurs [1][5][6]. Fungal spores, bacterial endotoxins, forage and storage mites, microbial toxins, peptidoglycans, proteases, pollen and plant debris, as well as inorganic particles trigger clinical signs of disease [5][6][7][8][9][10]. Several fungi (>50 species), especially Aspergillus fumigatus, have been widely recognised as significant risk factors for sEA [11][12][13]. Recent research by White and colleagues has uncovered the potential role of novel allergens including new species of fungi, mites, pollen, and arthropods, but also that of latex proteins [5], which hadn’t yet been clearly associated with the disease.
During disease exacerbation affected horses develop cough, nasal discharge and increased respiratory effort at rest [1][14][15], due to neutrophil recruitment, mucus plugging, bronchospasm and airway remodeling [16][17].
Severely asthmatic horses are usually managed through antigen avoidance and the use of corticosteroids and bronchodilators to reduce airway inflammation, bronchoconstriction and improve lung function [18]. However, some horses are unresponsive to corticosteroid treatment posing a challenge to clinicians. Thus, the identification of causal antigens and the development of antigen screening tests is fundamental and will enable a personalized treatment approach using specific immunotherapy [5][19].
Disease diagnosis is mostly based on history, clinical signs, and bronchoalveolar lavage fluid (BALF) differential cytology. Although lung function testing can accurately detect sEA, such equipment is unavailable to most field practitioners [1][14][20]. The genetic and immunological mechanisms associated with this disease are complex and heterogenous, implicating the activation of different inflammatory pathways [21][22][23]. Currently there is a need to better characterize the immune events leading to the occurrence and persistence of airway inflammation as this will help clinicians in determining the best treatment approach and in providing an accurate prognosis. Moreover, the development of novel ancillary diagnostic tests and therapeutic targets are required for early diagnosis of sEA and total resolution of airway inflammation in refractory cases.
Because sEA shares many similarities to its human counterpart, the horse is considered a good model for the study of the non-allergic and late on-set asthma phenotypes, since disease occurs naturally and sample collection can be easily performed [24]. Thus, further contributions to the disease’s characterization will benefit both horses and humans alike.

2. Genetic Background

Although sEA’s heritability has been shown in several horse breeds, and a familial aggregation has long been ascertain, external factors, such as environment, increase the likelihood of expressing the disease [3][21][22].
The chromosome region ECA13 has been associated with sEA in one family of Swiss Warmbloods, while region ECA15 has been implicated in a different family of the same breed. The inheritance mode differed between both families, being autosomal recessive in the first family and autosomal dominant in the second [25]. Additionally, in the first family of horses the Interleukin 4 receptor (IL-4R) gene and its neighboring regions in ECA13 appeared to contribute to disease in some individuals [26][27][28]. In humans, polymorphic differences in the Interleukin 4 receptor α chain (IL4Rα) gene play an important role in the development of asthma, since they induce the isotopic switch to immunoglobulin E (IgE) and the differentiation of T-helper type 2 (Th2) lymphocytes [29][30].
Racine and colleagues described an interaction between IL-4R and products of the SOCS5 gene, which may influence the molecular cascades involving nuclear factor (NF)-κB [31]. The gene coding for SOCS5 is located in the ECA15 chromosome region, and it is predominantly expressed by Th1 cells while further inhibiting Th2 differentiation. The inhibitory effect of SOCS5 on IL-4 signaling contributes to the non-Th2 cytokine profile observed in human non-allergic asthma [32], and may explain further similarities between both species.
In a genome wide association study (GWAS), the gene responsible for the TXNDC11 protein, also located in the ECA13 region, has been linked to sEA [33]. In humans, TXNDC11 controls the production of hydrogen peroxide in the respiratory epithelium [34], as well as the expression of MUC5AC mucin, which has been shown to play a significant role on airway hyperreactivity in mice [35]. In sEA-affected horses MUC5AC is upregulated, thus contributing to the mucus plugging observed in the disease [36].
The analysis of genomic copy number variants did not reveal any relevant variant regions which could be associated with the sEA, although a copy number loss was reported on chromosome 5 involving the gene NME7 [37]. The expression of this gene is necessary for ciliary function in the lungs and may be involved in sEA, since in knockout mice it induces primary ciliary dyskinesia [38]. Also, using RNA sequencing technique, a single point substitution was detected in the PACRG and RTTN genes in asthmatic horses, predictively altering their proteins, which are related to ciliary function [39].
In a gene set enrichment analysis of the bronchial epithelium after hay dust exposure, asthmatic horses presented upregulated genes of the E2F transcription factor family, which contribute to cell cycle regulation. Thus, asthmatic horses may suffer from impaired bronchial epithelial regeneration associated to subepithelial remodeling [40].
These recent studies have shown that the respiratory epithelium contributes to the immunological response observed in severely asthmatic horses.
Furthermore, an analysis of expression quantitative trait loci (eQTLs) allied with GWAS did not find a significant association between observed genetic variants and sEA, except for a disease-genetic variant in CLEC16A gene, which regulates gene expression of dexamethasone-induced protein (DEXI) [41]. This is of special importance in comparative pathology, as DEXI has also been reported in human asthma [42], although in horses it appears to not be a reliable indicator of sEA [41].
The identification and differential expression analysis of microRNAs (miRNAs) present in the serum of sEA-affected horses, showed a downregulation of miR-128 and miR-744. These findings suggest that a Th2/Th17 immunological response may characterize sEA [10][43].
Additionally, a recent work on Polish Konik horses aimed to detect the effects of inbreeding on sEA, however no effects were observed at the individual level [44].
Although most of these findings relate to certain families of Swiss Warmblood horses, they illustrate the complex genetic heterogeneity of sEA, which most likely results from the interaction of different genes. However, the use of such specific horse families and the likelihood of high variety of genetic background mechanisms contributing to the disease limits the application of these findings to the general equine population.

3. Immunological Phenotypes and Endotypes

Phenotype is the term used to describe the observable clinical characteristic of a disease, whereas endotype, a subclass of phenotype, refers to its molecular and genetic mechanism or treatment response [45].
As stated in the 2016 consensus, equine asthma (EA) is currently defined by two major phenotypes, which differ according to disease onset, clinical presentation and its severity—mild/moderate EA (mEA) and the already described sEA, which is the focus of this review [1]. However, phenotypes are insufficient when deciding upon the appropriate therapeutic management or determining the prognosis, which mainly depend on the immunological mechanisms of the disease.
Human asthma is usually considered to be a type 1 hypersensitivity, due to increased levels of IgE associated with a Th2 response, resulting in the recruitment of eosinophils into the airways [46]. However, an endotype which does not appear to be associated to a Th2 response has also been identified. As such, human asthma is divided into two major endotypes according to cytokine profile: Th2 and non-Th2 type asthma [47]. The Th2 type asthma is considered an allergic phenotype with the aforementioned eosinophil involvement and because its cytokine profile has been thoroughly described, several biomarkers are available for characterizing the disease and will be addressed bellow.
However, sEA is typically characterized by a neutrophilic response [1][48], and appears to not have the typical presentation of a type 1 hypersensitivity [49]. Although a Th2 cytokine profile has been described in sEA-affected horses, these animals do not display an early phase response [50][51]. However, a late phase response leading to neutrophilic bronchiolitis, associated with an increase in CD4+ T cells in the bronchoalveolar lavage fluid (BALF), has been described [50][52][53]. These features have led to the hypothesis that a type 3 hypersensitivity response, resulting in antibody-antigen complexes and activation of complement cascade, were involved in the disease’s immunology [54]. However, because sEA does not possess most of the features described in type 3 hypersensitivities, it is unlikely that this type of response accounts for the main immunological features of the disease [55].
Still the precise cytokine profile of sEA remains unclear, with a multitude of reports pointing to either a Th1, a Th2, a Th17 or a mixed mediated response. Table 1 illustrates the cytokines reported in sEA-affected horses.
Table 1. Cytokines reported in sEA-affected horses according to T helper subtype [10][43][49][52][56][57][58][59][60][61][62].
Th2 Th17 Th1/Th2 Th1/Th17 Th2/Th17 Undefined
↑ IL-4 1(r) ↑ CXCL13 2(r) ↑ IL-4 1(r) ↑ IL-1β1(r) ↓ miR-197 2(r) ↓ IFN-γ 1(r)
↑ IL-5 1(r)   ↑ IFN-γ 1(r) ↑ IL-8 1(r); 3(r); 3(p) ↑ miR-744 2(r) ↓ IL-4 1(r)
↓ IFN-γ 1(r)     ↑ IFN-γ 1(r) ↓ miR-26a 4(r) ↓ IL-5 1(r)
      ↑ TNF-α 1(r) ↑ miR-31 4(r) ↓ IL-13 1(r)
      ↑ IL-17 1(r) ↓ TNF-α 4(r)  
        ↑ IL-4R 4(r)  
1—BALF recovered cells; 2—Peripheral blood; 3—bronchial epithelial biopsy; 4—Lung tissue (post-mortem). r—RNA detected; p—protein detected. ↑—increased expression; ↓—decreased expression. IL—interleukin; IL-4R—interleukin 4 receptor; IFN-γ—gamma-interferon; TNF-α—tumor necrosis factor-α; miR—microRNA.
Using immunohistochemistry and in situ hybridization, the expression of IL-4 and IL-5 was observed in the BALF lymphocytes of sEA-affected horses [49][53]. However, the Th2 cytokine profile of these animals was accompanied by airway neutrophilia, but not eosinophilia.
Other authors have reported an increased expression of IL-1β, IL-8, gamma-interferon (IFN-γ), tumor necrosis factor (TNF)-α, and IL-17, mainly suggesting a Th1 and/or Th17 mixed mediated response [59][60][61].
Gene expression analysis of BALF cells and bronchial epithelium of severely asthmatic horses, using reverse transcription polymerase chain reaction (RT-PCR), revealed that the expression of IL-1β, IL-8, NF-κB and toll-like receptor (TLR)4 was upregulated in these animals. Furthermore, authors reported that these findings correlated with the neutrophil percentage detected in the BALF [59].
Ainsworth and colleagues reported that during remission severely asthmatic horses exhibited an increased expression of IL-13 and despite BALF neutrophilia no differences in cytokine expression were observed 24 h after environmental challenge. However, after 5 weeks of chronic exposure to aeroallergens asthmatic horses presented increased IFN-γ and IL-8 gene expression [60].
After antigen challenge, the BALF cells of sEA-affected horses showed elevated gene expression of IL-17, IL-8 and TLR4. Gene expression of IL-8 was also increased in the bronchial epithelium, and using immunohistochemistry was tracked to the ciliated epithelium of affected horses. Additionally, stimulated peripheral blood neutrophils of asthmatic horses incubated with lipopolysaccharide (LPS) and formyl-methionyl-leucine phenylalanine (fMLP), two potent pro-inflammatory agents associated with sEA, revealed upregulated gene expression of IL-17 and TLR4 [61].
The presence of a mixed Th1/Th2 cytokine profile, involving mediators such as IL-4 and IFN-γ, has also been reported [57][58]. Disease exacerbation, post-antigen challenge, was also accompanied by elevated expression of IL-1β, TNF-α, IL-8 and IFN-γ, and treatment with fluticasone decreased mRNA expression of TNF-α [57]. Similarly, horses diagnosed with summer pasture-associated sEA developed disease exacerbation during the summer months, with increased expression of IL-13 and IFN-γ by BALF lymphocytes and CD4+ lymphocytes from peripheral blood. Furthermore, during disease remission, in the winter, these animals exhibited increased IL-4 mRNA expression [58].
The possibility of a mixed Th2/Th17 response has also been postulated [43], associated with a dysregulated Th17 cell differentiation pathway [62]. Eleven differentially expressed miRNAs (DEmiRs) were reported in the serum of asthmatic horses, compared to healthy individuals. Also, a shift towards the maturation of Th2 cells was proposed, supported by decreased levels of miR-128, which in association with decreased miR-197 and increased levels of miR-744 affects the maturation of Th cells towards a Th17 profile [43].
The analysis of the miRNAs and mRNA found in the lung tissue of sEA-affected horses supports the hypothesis of a Th17 mediated response, but also of a Th2 immune response [62]. Additionally, the upregulated miRNAs miR-142-3p and miR-223 found in asthmatic horses are also associated with severe neutrophilic asthma in humans, and with increased expression of IL-1β, IL-6 and IL-8 [63] cytokines, some of which have been associated with sEA [57][59].
Contrarily, Kleiber and colleagues reported neither a specific Th1 nor a Th2 specific response, but a downregulation of expressed cytokines (IL-4, IL-5, IL-13 and IFN-γ) in the CD4 and CD8 populations of the peripheral blood and BALF of sEA-affected horses [56], which could implicate the involvement of other pathways in the disease.
Thus, the reported results may reflect the heterogeneity of the cytokine profile involved in sEA and may imply the existence of different disease endotypes. However the interpretation of these results must necessarily take into consideration the described methodologies of the above-mentioned studies. For example, cytokine expression was investigated using distinct samples, namely BALF, bronchial and lung tissue, as well as peripheral blood. As such, results may not only reflect the inflammatory response of the examined cells, but also differences between local and systemic inflammatory responses.
With the development of transcriptomics, novel techniques for assessing the existence and relative prevalence of several RNA species have been introduced to the scientific community. This is portrayed in the reported methodologies of the aforementioned studies, where recent experiments sequence mRNA and miRNA, contrasting with the less comprehensive/detailed methods, such as traditional targeted immunohistochemistry, in situ hybridization and RT-PCR.
Additionally, the experimental design of most studies involved the exacerbation of the disease by exposing the asthmatic horse to an intense pro-inflammatory environment, using hay dust and/or by stabling the affected horses. It cannot be excluded that the experimental induction of airway inflammation may interfere to some extent with the expressed cytokine profile, especially considering individual susceptibilities to specific allergens. Therefore, this factor also needs to be taken into account when interpreting reported results.
As in human asthma, it is highly likely that sEA possesses multiple endotypes, and considering the neutrophil recruitment observed in affected horses, a Th17 mediated response is probably part of the inflammatory pathways involved in this disease. Nevertheless, more encompassing studies involving genomics, transcriptomics and proteomics are required to better define the cytokine profile of sEA and to determine therapeutic targets in affected horses, and although further confirmation is required, the reported DEmiRs may constitute novel therapeutic targets for sEA.
Severely asthmatic horses may also present an altered response to allergens, since ex vivo and in vivo hay dust stimulation revealed upregulation of several genes participating in the inflammation [10][64]. Pacholewska and colleagues reported an increased expression of CXCL13 chemokine [10] which may indicate a Th17 mediated response [65], but no evidence of a Th1 nor a Th2 response was found. Additionally, in a murine model of allergic airway inflammation increased expression of CXCL13 has been reported and its neutralization reduced allergic inflammation by decreasing lymphocytes, eosinophils, as well as the recruitment of CXCR5-bearing cells [66]. Accordingly, in humans, IL-17 expression has been associated with severe neutrophilic asthma and in horses this cytokine is responsible for the activation and persistence of neutrophils in the airways. Also, IL-17 was shown to be associated with reduced response to corticosteroids, with post-treatment persistence of IL-8 [67][68].
The described heterogeneity also occurs in human asthma, where one could consider the existence of three distinct phenotypes: allergic asthma, non-allergic asthma and late-onset asthma [22]. These phenotypes may also be applicable when describing sEA. In this sense the allergic asthma phenotype would be characterized by a Th2 mediated response and usually associated with other allergic diseases. In general, horses affected with sEA may also suffer from other allergic diseases such as insect bite hypersensitivity or atopy [69][70]. Interestingly, Lo Feudo and colleagues have reported the presence of a type 1 hypersensitivity in sEA-affected horses in response to intradermal allergen test, which may further support the hypothesis of an allergic phenotype [71]. Additionally, the use of skin prick tests has previously been used to identify allergic sensitization in severely asthmatic horses [19]. Similarly, evidence of a type 1 hypersensitivity to different allergens has also been described in severely asthmatic horses using allergen inhalation [72].
The non-allergic phenotype in humans is usually associated with the presence of neutrophils in the airways, a hallmark of sEA. This phenotype also reflects the involvement of a Th1 and of a Th17 response [62], which has also been described as contributing to the immune response in asthmatic horses.
Finally, the late-onset asthma is age-associated and also occurs in sEA where affected individuals are mature adults [2][73]. This age association is thought to be the consequence of immunosenescence and inflammaging, which describe the immune and inflammatory modifications observed in geriatric patients [74][75][76], a subject extensively revised by Bullone et al. [75]. Immunosenescence is essentially a disfunction of the immune system. In horses it is usually characterized by a dysregulation of adaptative immunity associated with a lower proliferative response of T lymphocytes [77], and a decrease in mean percentage of regulatory T cells [78].
On the other hand, the term inflammaging defines the chronic inflammatory state observed in older individuals accompanied by an increased expression of inflammatory cytokines [74]. A correlation between age and IL-6 has also been described in healthy geriatric horses [79]. Also, compared to young adults, geriatric horses with colitis had higher levels of IL-6 and TNF-α [80]. It has also been described that older horses exhibit increased expression of IL-1β, IL-15, IL-18, IFN-γ and TNF-α mRNA [77][81][82]. These studies confirm that inflammaging and immunosenescence occur in geriatric horses both systemically and locally [74], and are most likely involved in the immunology of sEA, although further research is needed to clarify the age-associated changes and how they affect airway inflammatory response.
The reported differences in the immunological pathways contributing to sEA illustrate the complexity of this disease and suggest the existence of several endotypes, which converge into the same clinical phenotype. One must also consider that the methodological differences of the above mentioned studies, such as time of sample collection, natural vs. stimulated inflammatory response and duration of the disease, may have contributed to the reported variations. It is therefore fundamental that holistic studies, encompassing more exhaustive and complementary approaches, and preferably large multi-center studies can be performed to unravel sEA’s different immunological responses.

References

  1. Couëtil, L.L.; Cardwell, J.M.; Gerber, V.; Lavoie, J.P.; Léguillette, R.; Richard, E.A. Inflammatory Airway Disease of Horses-Revised Consensus Statement. J. Vet. Intern. Med. 2016, 30, 503–515.
  2. Hotchkiss, J.W.; Reid, S.W.J.; Christley, R.M. A survey of horse owners in Great Britain regarding horses in their care. Part 1: Horse demographic characteristics and management. Equine Vet. J. 2007, 39, 294–300.
  3. Ramseyer, A.; Gaillard, C.; Burger, D.; Straub, R.; Jost, U.; Boog, C.; Marti, E.; Gerber, V. Effects of Genetic and Environmental Factors on Chronic Lower Airway Disease in Horses. J. Vet. Intern. Med. 2007, 21, 149–156.
  4. Pirie, R.S.; Collie, D.D.S.; Dixon, P.M.; McGorum, B.C. Inhaled endotoxin and organic dust particulates have synergistic proinflammatory effects in equine heaves (organic dust-induced asthma). Clin. Exp. Allergy 2003, 33, 676–683.
  5. White, S.J.; Moore-Colyer, M.; Marti, E.; Hannant, D.; Gerber, V.; Coüetil, L.; Richard, E.A.; Alcocer, M. Antigen array for serological diagnosis and novel allergen identification in severe equine asthma. Sci. Rep. 2019, 9, 15171.
  6. Moore-Colyer, M.J.S.; Taylor, J.L.E.; James, R. The Effect of Steaming and Soaking on the Respirable Particle, Bacteria, Mould, and Nutrient Content in Hay for Horses. J. Equine Vet. Sci. 2016, 39, 62–68.
  7. Niedzwiedz, A.; Jaworski, Z.; Kubiak, K. Serum concentrations of allergen-specific IgE in horses with equine recurrent airway obstruction and healthy controls assessed by ELISA. Vet. Clin. Pathol. 2015, 44, 391–396.
  8. McGorum, B.C.; Ellison, J.; Cullen, R.T. Total and respirable airborne dust endotoxin concentrations in three equine management systems. Equine Vet. J. 1998, 30, 430–434.
  9. Klier, J.; Geis, S.; Steuer, J.; Geh, K.; Reese, S.; Fuchs, S.; Mueller, R.S.; Winter, G.; Gehlen, H. A comparison of nanoparticullate CpG immunotherapy with and without allergens in spontaneously equine asthma-affected horses, an animal model. Immun. Inflamm. Dis. 2018, 6, 81–96.
  10. Pacholewska, A.; Jagannathan, V.; Drögemüller, M.; Klukowska-Rötzler, J.; Lanz, S.; Hamza, E.; Dermitzakis, E.T.; Marti, E.; Leeb, T.; Gerber, V. Impaired cell cycle regulation in a natural equine model of asthma. PLoS ONE 2015, 10, e0136103.
  11. Pirie, R.S.; Dixon, P.M.; McGorum, B.C. Endotoxin contamination contributes to the pulmonary inflammatory and functional response to Aspergillus fumigatus extract inhalation in heaves horses. Clin. Exp. Allergy 2003, 33, 1289–1296.
  12. Schmallenbach, K.H.; Rahman, I.; Sasse, H.H.L.; Dixon, P.M.; Halliwell, R.E.W.; McGorum, B.C.; Crameri, R.; Miller, H.R.P. Studies on pulmonary and systemic Aspergillus fumigatus-specific IgE and IgG antibodies in horses affected with chronic obstructive pulmonary disease (COPD). Vet. Immunol. Immunopathol. 1998, 66, 245–256.
  13. Morán, G.; Folch, H.; Araya, O.; Burgos, R.; Barria, M. Detection of reaginic antibodies against Faenia rectivirgula from the serum of horses affected with Recurrent Airway Obstruction by an in vitro bioassay. Vet. Res. Commun. 2010, 34, 719–726.
  14. Tilley, P.; Sales Luis, J.P.; Branco Ferreira, M. Correlation and discriminant analysis between clinical, endoscopic, thoracic X-ray and bronchoalveolar lavage fluid cytology scores, for staging horses with recurrent airway obstruction (RAO). Res. Vet. Sci. 2012, 93, 1006–1014.
  15. Simões, J.; Sales Luís, J.; Tilley, P. Contribution of lung function tests to the staging of severe equine asthma syndrome in the field. Res. Vet. Sci. 2019, 123, 112–117.
  16. Pirie, R.S.; Collie, D.D.S.; Dixon, P.M.; McGorum, B.C. Evaluation of nebulised hay dust suspensions (HDS) for the diagnosis and investigation of heaves. 2: Effects of inhaled HDS on control and heaves horses. Equine Vet. J. 2002, 34, 337–342.
  17. Leclere, M.; Lavoie-Lamoureux, A.; Gélinas-Lymburner, É.; David, F.; Martin, J.G.; Lavoie, J.P. Effect of antigenic exposure on airway smooth muscle remodeling in an equine model of chronic asthma. Am. J. Respir. Cell Mol. Biol. 2011, 45, 181–187.
  18. Simões, J.; Sales Luís, J.P.; Tilley, P. Owner Compliance to an Environmental Management Protocol for Severe Equine Asthma Syndrome. J. Equine Vet. Sci. 2020, 87, 102937.
  19. Tilley, P.; Sales Luis, J.P.; Branco Ferreira, M. Comparison of Skin Prick Tests with In Vitro Allergy Tests in the Characterization of Horses with Recurrent Airway Obstruction. J. Equine Vet. Sci. 2012, 32, 719–727.
  20. Stucchi, L.; Ferrucci, F.; Bullone, M.; Dellacà, R.L.; Lavoie, J.P. Within-breath oscillatory mechanics in horses affected by severe equine asthma in exacerbation and in remission of the disease. Animals 2022, 12, 4.
  21. Gerber, V.; Baleri, D.; Klukowska-Rötzler, J.; Swinburne, J.E.; Dolf, G. Mixed inheritance of equine recurrent airway obstruction. J. Vet. Intern. Med. 2009, 23, 626–630.
  22. Marti, E.; Gerber, H.; Essich, G.; Oulehla, J.; Lazary, S. The genetic basis of equine allergic diseases 1. Chronic hypersensitivity bronchitis. Equine Vet. J. 1991, 23, 457–460.
  23. Couetil, L.; Cardwell, J.M.; Leguillette, R.; Mazan, M.; Richard, E.; Bienzle, D.; Bullone, M.; Gerber, V.; Ivester, K.; Lavoie, J.P.; et al. Equine Asthma: Current Understanding and Future Directions. Front. Vet. Sci. 2020, 7, 450.
  24. Bullone, M.; Lavoie, J.P. Asthma “of horses and men” - How can equine heaves help us better understand human asthma immunopathology and its functional consequences? Mol. Immunol. 2015, 66, 97–105.
  25. Swinburne, J.E.; Bogle, H.; Klukowska-Rötzler, J.; Drögemüller, M.; Leeb, T.; Temperton, E.; Dolf, G.; Gerber, V. A whole-genome scan for recurrent airway obstruction in Warmblood sport horses indicates two positional candidate regions. Mamm. Genome 2009, 20, 504–515.
  26. Klukowska-Rötzler, J.; Swinburne, J.E.; Drögemüller, C.; Dolf, G.; Janda, J.; Leeb, T.; Gerber, V. The interleukin 4 receptor gene and its role in recurrent airway obstruction in Swiss Warmblood horses. Anim. Genet. 2012, 43, 450–453.
  27. Shakhsi-Niaei, M.; Klukowska-Rötzler, J.; Drögemüller, C.; Swinburne, J.; Ehrmann, C.; Saftic, D.; Ramseyer, A.; Gerber, V.; Dolf, G.; Leeb, T. Replication and fine-mapping of a QTL for recurrent airway obstruction in European Warmblood horses. Anim. Genet. 2012, 43, 627–631.
  28. Jost, U.; Klukowska-Rötzler, J.; Dolf, G.; Swinburne, J.E.; Ramseyer, A.; Bugno, M.; Burger, D.; Blott, S.; Gerber, V. A region on equine chromosome 13 is linked to recurrent airway obstruction in horses. Equine Vet. J. 2007, 39, 236–241.
  29. Ober, C.; Leavitt, S.A.; Tsalenko, A.; Howard, T.D.; Hoki, D.M.; Daniel, R.; Newman, D.L.; Wu, X.; Parry, R.; Lester, L.A.; et al. Variation in the interleukin 4-receptor α gene confers susceptibility to asthma and atopy in ethnically diverse populations. Am. J. Hum. Genet. 2000, 66, 517–526.
  30. Youn, J.; Hwang, S.H.; Cho, C.S.; Min, J.K.; Kim, W.U.; Park, S.H.; Kim, H.Y. Association of the interleukin-4 receptor α variant Q576R with Th1/Th2 imbalance in connective tissue disease. Immunogenetics 2000, 51, 743–746.
  31. Racine, J.; Gerber, V.; Miskovic Feutz, M.; Riley, C.P.; Adamec, J.; Swinburne, J.E.; Couetil, L.L. Comparison of genomic and proteomic data in recurrent airway obstruction affected horses using ingenuity pathway analysis®. BMC Vet. Res. 2011, 7, 48.
  32. Seki, Y.I.; Hayashi, K.; Matsumoto, A.; Seki, N.; Tsukada, J.; Ransom, J.; Naka, T.; Kishimoto, T.; Yoshimura, A.; Kubo, M. Expression of the suppressor of cytokine signaling-5 (SOCS5) negatively regulates IL-4-dependent STAT6 activation and Th2 differentiation. Proc. Natl. Acad. Sci. USA 2002, 99, 13003–13008.
  33. Schnider, D.; Rieder, S.; Leeb, T.; Gerber, V.; Neuditschko, M. A genome-wide association study for equine recurrent airway obstruction in European Warmblood horses reveals a suggestive new quantitative trait locus on chromosome 13. Anim. Genet. 2017, 48, 691–693.
  34. Pacquelet, S.; Lehmann, M.; Luxen, S.; Regazzoni, K.; Frausto, M.; Noack, D.; Knaus, U.G. Inhibitory action of NoxA1 on dual oxidase activity in airway cells. J. Biol. Chem. 2008, 283, 24649–24658.
  35. Evans, C.M.; Raclawska, D.S.; Ttofali, F.; Liptzin, D.R.; Fletcher, A.A.; Harper, D.N.; McGing, M.A.; McElwee, M.M.; Williams, O.W.; Sanchez, E.; et al. The polymeric mucin Muc5ac is required for allergic airway hyperreactivity. Nat. Commun. 2015, 6, 6281.
  36. Gerber, V.; Robinson, N.E.; Venta, P.J.; Rawson, J.; Jefcoat, A.M.; Hotchkiss, J.A. Mucin genes in horse airways: MUC5AC, but not MUC2, may play a role in recurrent airway obstruction. Equine Vet. J. 2003, 35, 252–257.
  37. Ghosh, S.; Das, P.J.; McQueen, C.M.; Gerber, V.; Swiderski, C.E.; Lavoie, J.P.; Chowdhary, B.P.; Raudsepp, T. Analysis of genomic copy number variation in equine recurrent airway obstruction (heaves). Anim. Genet. 2016, 47, 334–344.
  38. Šedová, L.; Buková, I.; Bažantová, P.; Petrezsélyová, S.; Prochazka, J.; Školníková, E.; Zudová, D.; Včelák, J.; Makovický, P.; Bendlová, B.; et al. Semi-lethal primary ciliary dyskinesia in rats lacking the nme7 gene. Int. J. Mol. Sci. 2021, 22, 3810.
  39. Tessier, L.; Côté, O.; Bienzle, D. Sequence variant analysis of RNA sequences in severe equine asthma. PeerJ 2018, 2018, e5759.
  40. Tessier, L.; Côté, O.; Clark, M.E.; Viel, L.; Diaz-Méndez, A.; Anders, S.; Bienzle, D. Gene set enrichment analysis of the bronchial epithelium implicates contribution of cell cycle and tissue repair processes in equine asthma. Sci. Rep. 2018, 8, 16408.
  41. Mason, V.C.; Schaefer, R.J.; McCue, M.E.; Leeb, T.; Gerber, V. eQTL discovery and their association with severe equine asthma in European Warmblood horses. BMC Genom. 2018, 19, 581.
  42. Ferreira, M.A.R.; Matheson, M.C.; Tang, C.S.; Granell, R.; Ang, W.; Hui, J.; Kiefer, A.K.; Duffy, D.L.; Baltic, S.; Danoy, P.; et al. Genome-wide association analysis identifies 11 risk variants associated with the asthma with hay fever phenotype. J. Allergy Clin. Immunol. 2014, 133, 1564–1571.
  43. Pacholewska, A.; Kraft, M.F.; Gerber, V.; Jagannathan, V. Differential expression of serum MicroRNAs supports CD4+ t cell differentiation into Th2/Th17 cells in severe equine asthma. Genes 2017, 8, 383.
  44. Borowska, A.; Wolska, D.; Niedzwiedz, A.; Borowicz, H.; Jaworski, Z.; Siemieniuch, M.; Szwaczkowski, T. Some genetic and environmental effects on equine asthma in polish konik horses. Animals 2021, 11, 2285.
  45. Kuruvilla, M.E.; Lee, F.E.H.; Lee, G.B. Understanding Asthma Phenotypes, Endotypes, and Mechanisms of Disease. Clin. Rev. Allergy Immunol. 2019, 56, 219–233.
  46. Holgate, S.T. Innate and adaptive immune responses in asthma. Nat. Med. 2012, 18, 673–683.
  47. Kuo, C.H.S.; Pavlidis, S.; Loza, M.; Baribaud, F.; Rowe, A.; Pandis, I.; Sousa, A.; Corfield, J.; Djukanovic, R.; Lutter, R.; et al. T-helper cell type 2 (Th2) and non-Th2 molecular phenotypes of asthma using sputum transcriptomics in U-BIOPRED. Eur. Respir. J. 2017, 49, 1602135.
  48. Rossi, H.; Virtala, A.M.; Raekallio, M.; Rahkonen, E.; Rajamäki, M.M.; Mykkänen, A. Comparison of tracheal wash and bronchoalveolar lavage cytology in 154 horses with and without respiratory signs in a referral hospital over 2009–2015. Front. Vet. Sci. 2018, 5, 61.
  49. Cordeau, M.E.; Joubert, P.; Dewachi, O.; Hamid, Q.; Lavoie, J.P. IL-4, IL-5 and IFN-γ mRNA expression in pulmonary lymphocytes in equine heaves. Vet. Immunol. Immunopathol. 2004, 97, 87–96.
  50. McGorum, B.C.; Dixon, P.M.; Halliwell, R.E.W. Phenotypic analysis of peripheral blood and bronchoalveolar lavage fluid lymphocytes in control and chronic obstructive pulmonary disease affected horses, before and after “natural (hay and straw) challenges. Vet. Immunol. Immunopathol. 1993, 36, 207–222.
  51. Deaton, C.M.; Deaton, L.; Jose-Cunilleras, E.; Vincent, T.L.; Baird, A.W.; Dacre, K.; Marlin, D.J. Early onset airway obstruction in response to organic dust in the horse. J. Appl. Physiol. 2007, 102, 1071–1077.
  52. Kleiber, C.; Grünig, G.; Jungi, T.; Schmucker, N.; Gerber, H.; Davis, W.C.; Straub, R. Phenotypic Analysis of Bronchoalveolar Lavage Fluid Lymphocytes in Horses with Chronic Pulmonary Disease. J. Vet. Med. Ser. A Physiol. Pathol. Clin. Med. 1999, 46, 177–184.
  53. Lavoie, J.P.; Maghni, K.; Desnoyers, M.; Taha, R.; Martin, J.G.; Hamid, Q.A. Neutrophilic airway inflammation in horses with heaves is characterized by a Th2-type cytokine profile. Am. J. Respir. Crit. Care Med. 2001, 164, 1410–1413.
  54. Moran, G.; Folch, H.; Henriquez, C.; Ortloff, A.; Barria, M. Reaginic antibodies from horses with Recurrent Airway Obstruction produce mast cell stimulation. Vet. Res. Commun. 2012, 36, 251–258.
  55. Felippe, M.J.B. Equine Clinical Immunology, 1st ed.; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2016; ISBN 9781119086512.
  56. Kleiber, C.; McGorum, B.C.; Horohov, D.W.; Pirie, R.S.; Zurbriggen, A.; Straub, R. Cytokine profiles of peripheral blood and airway CD4 and CD8 T lymphocytes in horses with recurrent airway obstruction. Vet. Immunol. Immunopathol. 2005, 104, 91–97.
  57. Giguère, S.; Viel, L.; Lee, E.; MacKay, R.J.; Hernandez, J.; Franchini, M. Cytokine induction in pulmonary airways of horses with heaves and effect of therapy with inhaled fluticasone propionate. Vet. Immunol. Immunopathol. 2002, 85, 147–158.
  58. Horohov, D.W.; Beadle, R.E.; Mouch, S.; Pourciau, S.S. Temporal regulation of cytokine mRNA expression in equine recurrent airway obstruction. Vet. Immunol. Immunopathol. 2005, 108, 237–245.
  59. Padoan, E.; Ferraresso, S.; Pegolo, S.; Castagnaro, M.; Barnini, C.; Bargelloni, L. Real time RT-PCR analysis of inflammatory mediator expression in recurrent airway obstruction-affected horses. Vet. Immunol. Immunopathol. 2013, 156, 190–199.
  60. Ainsworth, D.M.; Grünig, G.; Matychak, M.B.; Young, J.; Wagner, B.; Erb, H.N.; Antczak, D.F. Recurrent airway obstruction (RAO) in horses is characterized by IFN-γ and IL-8 production in bronchoalveolar lavage cells. Vet. Immunol. Immunopathol. 2003, 96, 83–91.
  61. Ainsworth, D.M.; Wagner, B.; Franchini, M.; Grünig, G.; Erb, H.N.; Tan, J.Y. Time-dependent alterations in gene expression of interleukin-8 in the bronchial epithelium of horses with recurrent airway obstruction. Am. J. Vet. Res. 2006, 67, 669–677.
  62. Hulliger, M.F.; Pacholewska, A.; Vargas, A.; Lavoie, J.P.; Leeb, T.; Gerber, V.; Jagannathan, V. An integrative mirna-mrna expression analysis reveals striking transcriptomic similarities between severe equine asthma and specific asthma endotypes in humans. Genes 2020, 11, 1143.
  63. Maes, T.; Cobos, F.A.; Schleich, F.; Sorbello, V.; Henket, M.; De Preter, K.; Bracke, K.R.; Conickx, G.; Mesnil, C.; Vandesompele, J.; et al. Asthma inflammatory phenotypes show differential microRNA expression in sputum. J. Allergy Clin. Immunol. 2016, 137, 1433–1446.
  64. Tessier, L.; Côté, O.; Clark, M.E.; Viel, L.; Diaz-Méndez, A.; Anders, S.; Bienzle, D. Impaired response of the bronchial epithelium to inflammation characterizes severe equine asthma. BMC Genom. 2017, 18, 708.
  65. Takagi, R.; Higashi, T.; Hashimoto, K.; Nakano, K.; Mizuno, Y.; Okazaki, Y.; Matsushita, S. B Cell Chemoattractant CXCL13 Is Preferentially Expressed by Human Th17 Cell Clones. J. Immunol. 2008, 181, 186–189.
  66. Baay-Guzman, G.J.; Huerta-Yepez, S.; Vega, M.I.; Aguilar-Leon, D.; Campillos, M.; Blake, J.; Benes, V.; Hernandez-Pando, R.; Teran, L.M. Role of CXCL13 in asthma: Novel therapeutic target. Chest 2012, 141, 886–894.
  67. Debrue, M.; Hamilton, E.; Joubert, P.; Lajoie-Kadoch, S.; Lavoie, J.P. Chronic exacerbation of equine heaves is associated with an increased expression of interleukin-17 mRNA in bronchoalveolar lavage cells. Vet. Immunol. Immunopathol. 2005, 105, 25–31.
  68. Murcia, R.Y.; Vargas, A.; Lavoie, J.P. The interleukin-17 induced activation and increased survival of equine neutrophils is insensitive to glucocorticoids. PLoS ONE 2016, 11, e0154755.
  69. Kehrli, D.; Jandova, V.; Fey, K.; Jahn, P.; Gerber, V. Multiple hypersensitivities including recurrent airway obstruction, insect bite hypersensitivity, and urticaria in 2 warmblood horse populations. J. Vet. Intern. Med. 2015, 29, 320–326.
  70. Lanz, S.; Brunner, A.; Graubner, C.; Marti, E.; Gerber, V. Insect Bite Hypersensitivity in Horses is Associated with Airway Hyperreactivity. J. Vet. Intern. Med. 2017, 31, 1877–1883.
  71. Lo Feudo, C.M.; Stucchi, L.; Alberti, E.; Conturba, B.; Zucca, E.; Ferrucci, F. Intradermal testing results in horses affected by mild-moderate and severe equine asthma. Animals 2021, 11, 2086.
  72. Klier, J.; Lindner, D.; Reese, S.; Mueller, R.S.; Gehlen, H. Comparison of Four Different Allergy Tests in Equine Asthma Affected Horses and Allergen Inhalation Provocation Test. J. Equine Vet. Sci. 2021, 102, 103433.
  73. Couëtil, L.L.; Ward, M.P. Analysis of risk factors for recurrent airway obstruction in North American horses: 1,444 Cases (1990-1999). J. Am. Vet. Med. Assoc. 2003, 223, 1645–1650.
  74. Hansen, S.; Baptiste, K.E.; Fjeldborg, J.; Horohov, D.W. A review of the equine age-related changes in the immune system: Comparisons between human and equine aging, with focus on lung-specific immune-aging. Ageing Res. Rev. 2015, 20, 11–23.
  75. Bullone, M.; Lavoie, J.P. The contribution of oxidative stress and inflamm-aging in human and equine asthma. Int. J. Mol. Sci. 2017, 18, 2612.
  76. Franceschi, C.; Bonafè, M.; Valensin, S.; Olivieri, F.; De Luca, M.; Ottaviani, E.; De Benedictis, G. Inflamm-aging. An evolutionary perspective on immunosenescence. Ann. N. Y. Acad. Sci. 2000, 908, 244–254.
  77. Adams, A.A.; Breathnach, C.C.; Katepalli, M.P.; Kohler, K.; Horohov, D.W. Advanced age in horses affects divisional history of T cells and inflammatory cytokine production. Mech. Ageing Dev. 2008, 129, 656–664.
  78. Robbin, M.G.; Wagner, B.; Noronha, L.E.; Antczak, D.F.; De Mestre, A.M. Subpopulations of equine blood lymphocytes expressing regulatory T cell markers. Vet. Immunol. Immunopathol. 2011, 140, 90–101.
  79. Suagee, J.K.; Corl, B.A.; Crisman, M.V.; Pleasant, R.S.; Thatcher, C.D.; Geor, R.J. Relationships between Body Condition Score and Plasma Inflammatory Cytokines, Insulin, and Lipids in a Mixed Population of Light-Breed Horses. J. Vet. Intern. Med. 2013, 27, 157–163.
  80. Sage, S.E.; Bedenice, D.; McKinney, C.A.; Long, A.E.; Pacheco, A.; Wagner, B.; Mazan, M.R.; Paradis, M.R. Assessment of the impact of age and of blood-derived inflammatory markers in horses with colitis. J. Vet. Emerg. Crit. Care 2021, 31, 779–787.
  81. McFarlane, D.; Holbrook, T.C. Cytokine dysregulation in aged horses and horses with pituitary pars intermedia dysfunction. J. Vet. Intern. Med. 2008, 22, 436–442.
  82. Hansen, S.; Sun, L.; Baptiste, K.E.; Fjeldborg, J.; Horohov, D.W. Age-related changes in intracellular expression of IFN-γ and TNF-α in equine lymphocytes measured in bronchoalveolar lavage and peripheral blood. Dev. Comp. Immunol. 2013, 39, 228–233.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 661
Revisions: 5 times (View History)
Update Date: 08 Apr 2022
1000/1000
ScholarVision Creations