Immunology2023; doi: 10.1111/imm.13745

Single-cell profiling of bronchoalveolar cells reveals a Th17 signature in neutrophilic severe equine asthma.

Abstract: Severe equine asthma (SEA) is a complex respiratory condition characterized by chronic airway inflammation. It shares many clinical and pathological features with human neutrophilic asthma, making it a valuable model for studying this condition. However, the immune mechanisms driving SEA have remained elusive. Although SEA has been primarily associated with a Th2 response, there have also been reports of Th1, Th17, or mixed-mediated responses. To uncover the elusive immune mechanisms driving SEA, we performed single-cell mRNA sequencing (scRNA-seq) on cryopreserved bronchoalveolar cells from 11 Warmblood horses, 5 controls and 6 with SEA. We identified six major cell types, including B cells, T cells, monocytes-macrophages, dendritic cells, neutrophils, and mast cells. All cell types exhibited significant heterogeneity, with previously identified and novel cell subtypes. Notably, we observed monocyte-lymphocyte complexes and detected a robust Th17 signature in SEA, with CXCL13 upregulation in intermediate monocytes. Asthmatic horses exhibited expansion of the B-cell population, Th17 polarization of the T-cell populations, and dysregulation of genes associated with T-cell function. Neutrophils demonstrated enhanced migratory capacity and heightened aptitude for neutrophil extracellular trap formation. These findings provide compelling evidence for a predominant Th17 immune response in neutrophilic SEA, driven by dysregulation of monocyte and T-cell genes. The dysregulated genes identified through scRNA-seq have potential as biomarkers and therapeutic targets for SEA and provide insights into human neutrophilic asthma.
Publication Date: 2023-12-28 PubMed ID: 38153159DOI: 10.1111/imm.13745Google Scholar: Lookup
The Equine Research Bank provides access to a large database of publicly available scientific literature. Inclusion in the Research Bank does not imply endorsement of study methods or findings by Mad Barn.
  • Journal Article

Summary

This research summary has been generated with artificial intelligence and may contain errors and omissions. Refer to the original study to confirm details provided. Submit correction.

The research article focuses on a detailed analysis of bronchoalveolar cells in horses suffering from severe equine asthma (SEA), revealing a predominant Th17 immune response and potentially providing new biomarkers and therapeutic targets for treating the condition.

Research Objective and Methodology

The researchers aimed to uncover the elusive immune mechanisms driving SEA, a complex respiratory condition characterized by chronic airway inflammation. The study uses SEA as a model for studying human neutrophilic asthma given the similar clinical and pathological features. For this, the researchers used single-cell mRNA sequencing (scRNA-seq) on bronchoalveolar cells from 11 Warmblood horses; five were controls and six were afflicted with SEA.

Findings of the Study

  • Through the procedure, the researchers identified six primary cell types: B cells, T cells, monocytes-macrophages, dendritic cells, neutrophils, and mast cells. Significant heterogeneity was found within all cell types, including previously identified and novel cell subtypes.
  • Notably, the researchers observed complexes formed by monocytes and lymphocytes and detected a strong Th17 signature in SEA. An upregulation in CXCL13, a gene responsible for controlling chemotaxis, was found in intermediate monocytes.
  • In horses with asthma, the researchers found an expansion of the B-cell population and Th17 polarization of the T-cell populations. They also found a dysregulation of genes associated with T-cell function.
  • The researchers also found that the neutrophils demonstrated enhanced migratory capacity and a heightened ability to form neutrophil extracellular traps, a component of the immune response.

Conclusion and Implications

The results reveal a predominant Th17 immune response in cases of neutrophilic SEA. This response is driven by the dysregulation of particular genes in monocytes and T-cells. The dysregulated genes identified through scRNA-seq potentially serve as biomarkers for SEA and possible targets for therapeutic intervention. Consequently, the findings of this study also shed light on the understanding and treatment of human neutrophilic asthma.

Cite This Article

APA
Sage SE, Leeb T, Jagannathan V, Gerber V. (2023). Single-cell profiling of bronchoalveolar cells reveals a Th17 signature in neutrophilic severe equine asthma. Immunology. https://doi.org/10.1111/imm.13745

Publication

ISSN: 1365-2567
NlmUniqueID: 0374672
Country: England
Language: English

Researcher Affiliations

Sage, Sophie E
  • Department of Clinical Veterinary Medicine, Vetsuisse Faculty, Swiss Institute of Equine Medicine, University of Bern, Bern, Switzerland.
Leeb, Tosso
  • Institute of Genetics, Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, Switzerland.
Jagannathan, Vidhya
  • Institute of Genetics, Vetsuisse Faculty, Institute of Genetics, University of Bern, Bern, Switzerland.
Gerber, Vinzenz
  • Department of Clinical Veterinary Medicine, Vetsuisse Faculty, Swiss Institute of Equine Medicine, University of Bern, Bern, Switzerland.

Grant Funding

  • 31003A-162548/1 / Swiss National Science Foundation

References

This article includes 78 references
  1. Couu00ebtil LLL, Cardwell JMM, Gerber V, Lavoie J-PP, Lu00e9guillette R, Richard EAA. Inflammatory airway disease of horses-revised consensus Statement. J Vet Intern Med. 2016;30(2):503-515. https://doi.org/10.1111/jvim.13824
  2. Laumen E, Doherr MG, Gerber V. Relationship of horse owner assessed respiratory signs index to characteristics of recurrent airway obstruction in two warmblood families. Equine Vet J. 2010;42(2):142-148. https://doi.org/10.2746/042516409X479586
  3. Ramseyer A, Gaillard C, Burger D, Straub R, Jost U, Boog C, et al. Effects of genetic and environmental factors on chronic lower airway disease in horses. J Vet Intern Med. 2007;21(1):149-156. https://doi.org/10.1111/j.1939-1676.2007.tb02941.x
  4. 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(6):691-693. https://doi.org/10.1111/age.12583
  5. Eder C, Curik I, Brem G, Crameri R, Bodo I, Habe F, et al. Influence of environmental and genetic factors on allergen-specific immunoglobulin-E levels in sera from Lipizzan horses. Equine Vet J. 2010;33(7):714-720. https://doi.org/10.2746/042516401776249264
  6. Bond S, Lu00e9guillette R, Richard EA, Couetil L, Lavoie JP, Martin JG, et al. Equine asthma: integrative biologic relevance of a recently proposed nomenclature. J Vet Intern Med. 2018;32(6):2088-2098. https://doi.org/10.1111/jvim.15302
  7. Woodrow JS, Sheats MK, Cooper B, Bayless R. Asthma: the use of animal models and their translational utility. Cell. 2023;12(7):1091. https://doi.org/10.3390/cells12071091
  8. Sheats MK, Davis KU, Poole JA. Comparative review of asthma in farmers and horses. Curr Allergy Asthma Rep. 2019;19(11):50. https://doi.org/10.1007/s11882-019-0882-2
  9. Lange-Consiglio A, Stucchi L, Zucca E, Lavoie JP, Cremonesi F, Ferrucci F. Insights into animal models for cell-based therapies in translational studies of lung diseases: is the horse with naturally occurring asthma the right choice? Cytotherapy. 2019;21(5):525-534. https://doi.org/10.1016/j.jcyt.2019.02.010
  10. Simu00f5es J, Batista M, Tilley P. The immune mechanisms of severe equine asthma-current understanding and what is missing. Animals. 2022;12(6):744. https://doi.org/10.3390/ani12060744
  11. 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(1):320-326. https://doi.org/10.1111/jvim.12473
  12. 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(6):1877-1883. https://doi.org/10.1111/jvim.14817
  13. 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(1-2):25-31. https://doi.org/10.1016/j.vetimm.2004.12.013
  14. Korn A, Miller D, Dong L, Buckles EL, Wagner B, Ainsworth DM. Differential 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 response. PLoS One. 2015;10(11):e0142622. https://doi.org/10.1371/journal.pone.0142622
  15. Pacholewska A, Kraft MF, Gerber V, Jagannathan V. Differential expression of serum MicroRNAs supports CD4 + t cell differentiation into Th2/Th17 cells in severe equine asthma. Genes (Basel). 2017;8(12):383. https://doi.org/10.3390/genes8120383
  16. Hulliger MF, Pacholewska A, Vargas A, Lavoie JP, Leeb T, Gerber V, et al. An integrative miRNA-mRNA expression analysis reveals striking transcriptomic similarities between severe equine asthma and specific asthma endotypes in humans. Genes (Basel). 2020;11(10):1143. https://doi.org/10.3390/genes11101143
  17. Pacholewska A, Jagannathan V, Dru00f6gemu00fcller M, Klukowska-Ru00f6tzler J, Lanz S, Hamza E, et al. Impaired cell cycle regulation in a natural equine model of asthma. PLoS One. 2015;10(8):e0136103. https://doi.org/10.1371/journal.pone.0136103
  18. Gressler AE, Lu00fcbke S, Wagner B, Arnold C, Lohmann KL, Schnabel CL. Comprehensive flow cytometric characterization of bronchoalveolar lavage cells indicates comparable phenotypes between asthmatic and healthy horses but functional lymphocyte differences. Front Immunol. 2022;13:1-17. https://doi.org/10.3389/fimmu.2022.896255
  19. Sage SE, Nicholson P, Peters LM, Leeb T, Jagannathan V, Gerber V. Single-cell gene expression analysis of cryopreserved equine bronchoalveolar cells. Front Immunol. 2022;13:1-18. https://doi.org/10.3389/fimmu.2022.929922
  20. Liu L, Zhang X, Liu Y, Zhang L, Zheng J, Wang J, et al. Chitinase-like protein YKL-40 correlates with inflammatory phenotypes, anti-asthma responsiveness and future exacerbations. Respir Res. 2019;20(1):1-12. https://doi.org/10.1186/s12931-019-1051-9
  21. Cremades-Jimeno L, de Pedro Mu00c1, Lu00f3pez-Ramos M, Sastre J, Mu00ednguez P, Fernu00e1ndez IM, et al. Prioritizing molecular biomarkers in asthma and respiratory allergy using systems biology. Front Immunol. 2021;12:1004. https://doi.org/10.3389/fimmu.2021.640791
  22. Alevy YG, Patel AC, Romero AG, Patel DA, Tucker J, Roswit WT, et al. IL-13-induced airway mucus production is attenuated by MAPK13 inhibition. J Clin Invest. 2012;122(12):4555-4568. https://doi.org/10.1172/JCI64896
  23. Bao C, Liu C, Liu Q, Hua L, Hu J, Li Z, et al. Liproxstatin-1 alleviates LPS/IL-13-induced bronchial epithelial cell injury and neutrophilic asthma in mice by inhibiting ferroptosis. Int Immunopharmacol. 2022;109:108770. https://doi.org/10.1016/j.intimp.2022.108770
  24. Sprenkeler EGG, Zandstra J, van Kleef ND, Goetschalckx I, Verstegen B, Aarts CEM, et al. S100A8/A9 is a marker for the release of neutrophil extracellular traps and induces neutrophil activation. Cell. 2022;11(2):236. https://doi.org/10.3390/cells11020236
  25. Jiang S, Park DW, Tadie J-M, Gregoire M, Deshane J, Pittet JF, et al. Human Resistin promotes neutrophil proinflammatory activation and neutrophil extracellular trap formation and increases severity of acute lung injury. J Immunol. 2014;192(10):4795-4803. https://doi.org/10.4049/jimmunol.1302764
  26. Zhu L, Zeng D, Lei X, Huang J, Deng YF, Ji YB, et al. KLF2 regulates neutrophil migration by modulating CXCR1 and CXCR2 in asthma. Biochim Biophys Acta Mol Basis Dis. 2020;1866(12):165920. https://doi.org/10.1016/j.bbadis.2020.165920
  27. Le A, Wu Y, Liu W, Wu C, Hu P, Zou J, et al. MiR-144-induced KLF2 inhibition and NF-kappaB/CXCR1 activation promote neutrophil extracellular trap-induced transfusion-related acute lung injury. J Cell Mol Med. 2021;25(14):6511-6523. https://doi.org/10.1111/jcmm.16650
  28. Leemans JC, te Velde AA, Florquin S, Bennink RJ, de Bruin K, van Lier u0154AW, et al. The epidermal growth factor-seven transmembrane (EGF-TM7) receptor CD97 is required for neutrophil migration and host defense. J Immunol. 2004;172(2):1125-1131. https://doi.org/10.4049/jimmunol.172.2.1125
  29. Majchrzak-Gorecka M, Majewski P, Grygier B, Murzyn K, Cichy J. Secretory leukocyte protease inhibitor (SLPI), a multifunctional protein in the host defense response. Cytokine Growth Factor Rev. 2016;28:79-93. https://doi.org/10.1016/j.cytogfr.2015.12.001
  30. Prince LR, Prosseda SD, Higgins K, Carlring J, Prestwich EC, Ogryzko NV, et al. NR4A orphan nuclear receptor family members, NR4A2 and NR4A3, regulate neutrophil number and survival. Blood. 2017;130(8):1014-1025. https://doi.org/10.1182/blood-2017-03-770164
  31. Cardenas EI, Che KF, Konradsen JR, Bao A, Lindu00e9n A. IL-26 in asthma and COPD. Expert Rev Respir Med. 2022;16(3):293-301. https://doi.org/10.1080/17476348.2022.2045197
  32. Chen X, Khalid K, Chen D, Qiu C. Serum levels of olfactomedin 4: a biomarker for asthma control state in asthmatics. Ann Transl Med. 2020;8(7):494. https://doi.org/10.21037/atm.2020.03.213
  33. Al Mutairi SS, Mojiminiyi OA, Shihab-Eldeen A, Al Rammah T, Abdella N. Putative roles of circulating resistin in patients with asthma, COPD and cigarette smokers. Dis Markers. 2011;31(1):1-7. https://doi.org/10.3233/DMA-2011-0793
  34. Mailer RKW, Joly A-L, Liu S, Elias S, Tegner J, Andersson J. IL-1u03b2 promotes Th17 differentiation by inducing alternative splicing of FOXP3. Sci Rep. 2015;5(1):14674. https://doi.org/10.1038/srep14674
  35. Gruarin P, Maglie S, De Simone M, Hu00e4ringer B, Vasco C, Ranzani V, et al. Eomesodermin controls a unique differentiation program in human IL-10 and IFN-u03b3 coproducing regulatory T cells. Eur J Immunol. 2019;49(1):96-111. https://doi.org/10.1002/eji.201847722
  36. Evans MD, Esnault S, Denlinger LC, Jarjour NN. Sputum cell IL-1 receptor expression level is a marker of airway neutrophilia and airflow obstruction in asthmatic patients. J Allergy Clin Immunol. 2018;142(2):415-423. https://doi.org/10.1016/j.jaci.2017.09.035
  37. Akitsu A, Iwakura Y. Interleukin-17-producing u03b3u03b4 T (u03b3u03b417) cells in inflammatory diseases. Immunology. 2018;155(4):418-426. https://doi.org/10.1111/imm.12993
  38. Seo H, Chen J, Gonzu00e1lez-Avalos E, Samaniego-Castruita D, das A, Wang YH, et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8 + T cell exhaustion. Proc Natl Acad Sci U S A. 2019;116(25):12410-12415. https://doi.org/10.1073/pnas.1905675116
  39. Evrard M, Wynne-Jones E, Peng C, Kato Y, Christo SN, Fonseca R, et al. Sphingosine 1-phosphate receptor 5 (S1PR5) regulates the peripheral retention of tissue-resident lymphocytes. J Exp Med. 2022;219(1):e20210116. https://doi.org/10.1084/jem.20210116
  40. Hirota JA, Hiebert PR, Gold M, Wu D, Graydon C, Smith JA, et al. Granzyme B deficiency exacerbates lung inflammation in mice after acute lung injury. Am J Respir Cell Mol Biol. 2013;49(3):453-462. https://doi.org/10.1165/rcmb.2012-0512OC
  41. Ingram JT, Yi JS, Zajac AJ. Exhausted CD8 T cells downregulate the IL-18 receptor and become unresponsive to inflammatory cytokines and bacterial Co-infections. PLoS Pathog. 2011;7(9):e1002273. https://doi.org/10.1371/journal.ppat.1002273
  42. Nguyen KD, Fohner A, Booker JD, Dong C, Krensky AM, Nadeau KC. XCL1 enhances regulatory activities of CD4+CD25highCD127low/u2212 T cells in human allergic asthma. J Immunol. 2008;181(8):5386-5395. https://doi.org/10.4049/jimmunol.181.8.5386
  43. Nair MPN, Schwartz SA, Wu K, Kronfol Z. Effect of neuropeptide Y on natural killer activity of normal human lymphocytes. Brain Behav Immun. 1993;7(1):70-78. https://doi.org/10.1006/brbi.1993.1007
  44. Brand HK, Ahout IML, de Ridder D, van Diepen A, Li Y, Zaalberg M, et al. Olfactomedin 4 serves as a marker for disease severity in pediatric respiratory syncytial virus (RSV) infection. PLoS One. 2015;10(7):e0131927. https://doi.org/10.1371/journal.pone.0131927
  45. Gong F, Li R, Zheng X, Chen W, Zheng Y, Yang Z, et al. OLFM4 regulates lung epithelial cell function in sepsis-associated ARDS/ALI via LDHA-mediated NF-u03baB signaling. J Inflamm Res. 2021;14(12):7035-7051. https://doi.org/10.2147/JIR.S335915
  46. Bozinovski S, Cross M, Vlahos R, Jones JE, Hsuu K, Tessier PA, et al. S100A8 chemotactic protein is abundantly increased, but only a minor contributor to LPS-induced, steroid resistant neutrophilic lung inflammation in vivo. J Proteome Res. 2005;4(1):136-145. https://doi.org/10.1021/pr049829t
  47. Wjst M. Exome variants associated with asthma and allergy. Sci Rep. 2022;12(1):21028. https://doi.org/10.1038/s41598-022-24960-6
  48. Park SY, Jing X, Gupta D, Dziarski R. Peptidoglycan recognition protein 1 enhances experimental asthma by promoting Th2 and Th17 and limiting regulatory T cell and plasmacytoid dendritic cell responses. J Immunol. 2013;190(7):3480-3492. https://doi.org/10.4049/jimmunol.1202675
  49. Gao P, Tang K, Wang M, Yang Q, Xu Y, Wang J, et al. Pentraxin levels in non-eosinophilic versus eosinophilic asthma. Clin Exp Allergy. 2018;48(8):981-989. https://doi.org/10.1111/cea.13168
  50. Ramery E, Fievez L, Fraipont A, Bureau F, Lekeux P. Characterization of pentraxin 3 in the horse and its expression in airways. Vet Res. 2010;41(2):18. https://doi.org/10.1051/vetres/2009066
  51. Kotsiou OS, Papagiannis D, Papadopoulou R, Gourgoulianis KI. Calprotectin in lung diseases. Int J Mol Sci. 2021;22(4):1706. https://doi.org/10.3390/ijms22041706
  52. Lin Q, Ni H, Zhong J, Zheng Z, Nie H. Identification of hub genes and potential biomarkers of neutrophilic asthma: evidence from a bioinformatics analysis. J Asthma. 2023;60(2):348-359. https://doi.org/10.1080/02770903.2022.2051544
  53. Ai X, Shen H, Wang Y, Zhuang J, Zhou Y, Niu F, et al. Developing a diagnostic model to predict the risk of asthma based on ten macrophage-related gene signatures. Biomed Res Int. 2022;2022:1-14. https://doi.org/10.1155/2022/3439010
  54. Wang Q, Liu S, Min J, Yin M, Zhang Y, Zhang Y, et al. CCL17 drives fibroblast activation in the progression of pulmonary fibrosis by enhancing the TGF-u03b2/Smad signaling. Biochem Pharmacol. 2023;210:115475. https://doi.org/10.1016/j.bcp.2023.115475
  55. Debien E, Mayol K, Biajoux V, Daussy C, de Aguero MG, Taillardet M, et al. S1PR5 is pivotal for the homeostasis of patrolling monocytes. Eur J Immunol. 2013;43(6):1667-1675. https://doi.org/10.1002/eji.201343312
  56. Hoek KL, Greer MJ, McClanahan KG, Nazmi A, Piazuelo MB, Singh K, et al. Granzyme B prevents aberrant IL-17 production and intestinal pathogenicity in CD4+ T cells. Mucosal Immunol. 2021;14(5):1088-1099. https://doi.org/10.1038/s41385-021-00427-1
  57. Deng J, Yu X-Q, Wang P-H. Inflammasome activation and Th17 responses. Mol Immunol. 2019;107:142-164. https://doi.org/10.1016/j.molimm.2018.12.024
  58. Arredouani MS, Franco F, Imrich A, Fedulov A, Lu X, Perkins D, et al. Scavenger receptors SR-AI/II and MARCO limit pulmonary dendritic cell migration and allergic airway inflammation. J Immunol. 2007;178(9):5912-5920. https://doi.org/10.4049/jimmunol.178.9.5912
  59. Hanschmann E-M, Berndt C, Hecker C, Garn H, Bertrams W, Lillig CH, et al. Glutaredoxin 2 reduces asthma-like acute airway inflammation in mice. Front Immunol. 2020;11(November):1-11. https://doi.org/10.3389/fimmu.2020.561724
  60. Branchett WJ, Cook J, Oliver RA, Bruno N, Walker SA, Stu00f6lting H, et al. Airway macrophage-intrinsic TGF-u03b21 regulates pulmonary immunity during early-life allergen exposure. J Allergy Clin Immunol. 2021;147(5):1892-1906. https://doi.org/10.1016/j.jaci.2021.01.026
  61. McDonough JE, Ahangari F, Li Q, Jain S, Verleden SE, Herazo-Maya J, et al. Transcriptional regulatory model of fibrosis progression in the human lung. JCI Insight. 2019;4(22):e131597. https://doi.org/10.1172/jci.insight.131597
  62. Zhang D-W, Ye J-J, Sun Y, Ji S, Kang JY, Wei YY, et al. CD19 and POU2AF1 are potential immune-related biomarkers involved in the emphysema of COPD: on multiple microarray analysis. J Inflamm Res. 2022;15(April):2491-2507. https://doi.org/10.2147/JIR.S355764
  63. Di Mauro S, Scamporrino A, Fruciano M, Filippello A, Fagone E, Gili E, et al. Circulating coding and long non-coding RNAs as potential biomarkers of idiopathic pulmonary fibrosis. Int J Mol Sci. 2020;21(22):8812. https://doi.org/10.3390/ijms21228812
  64. Xie Y, Abel PW, Casale TB, Tu Y. TH17 cells and corticosteroid insensitivity in severe asthma. J Allergy Clin Immunol. 2022;149(2):467-479. https://doi.org/10.1016/J.JACI.2021.12.769
  65. Baay-Guzman GJ, Huerta-Yepez S, Vega MI, Aguilar-Leon D, Campillos M, Blake J, et al. Role of CXCL13 in asthma: novel therapeutic target. Chest. 2012;141(4):886-894. https://doi.org/10.1378/chest.11-0633
  66. Alturaiki W. Elevated plasma levels of CXCL13 chemokine in Saudi patients with asthma exacerbation. Cureus. 2022;14(1):1-7. https://doi.org/10.7759/cureus.21142
  67. Takagi R, Higashi T, Hashimoto K, Nakano K, Mizuno Y, Okazaki Y, et al. B cell chemoattractant CXCL13 is preferentially expressed by human Th17 cell clones. J Immunol. 2008;181(1):186-189. https://doi.org/10.4049/jimmunol.181.1.186
  68. Al-Kufaidy R, Vazquez-Tello A, BaHammam AS, Al-Muhsen S, Hamid Q, Halwani R. IL-17 enhances the migration of B cells during asthma by inducing CXCL13 chemokine production in structural lung cells. J Allergy Clin Immunol. 2017;139(2):696-699.e5. https://doi.org/10.1016/j.jaci.2016.07.037
  69. Roberts CA, Dickinson AK, Taams LS. The interplay between monocytes/macrophages and CD4+ T cell subsets in rheumatoid arthritis. Front Immunol. 2015;6:571. https://doi.org/10.3389/fimmu.2015.00571
  70. Pang Y, Du X, Xu X, Wang M, Li Z. Monocyte activation and inflammation can exacerbate Treg/Th17 imbalance in infants with neonatal necrotizing enterocolitis. Int Immunopharmacol. 2018;59:354-360. https://doi.org/10.1016/j.intimp.2018.04.026
  71. Evans HG, Gullick NJ, Kelly S, Pitzalis C, Lord GM, Kirkham BW, et al. In vivo activated monocytes from the site of inflammation in humans specifically promote Th17 responses. Proc Natl Acad Sci U S A. 2009;106(15):6232-6237. https://doi.org/10.1073/pnas.0808144106
  72. Riihimu00e4ki M, Fegraeus K, Nordlund J, Waern I, Wernersson S, Akula S, et al. Single cell transcriptomics delineates the immune-cell landscape in equine lower airways and reveals upregulation of the FKBP5 gene in horses with asthmar. Sci Rep. 2023;13:16261. https://doi.org/10.21203/rs.3.rs-2768703/v1
  73. Burel JG, Pomaznoy M, Lindestam Arlehamn CS, Weiskopf D, da Silva Antunes R, Jung Y, et al. Circulating T cell-monocyte complexes are markers of immune perturbations. Elife. 2019;8:1-21. https://doi.org/10.7554/eLife.46045
  74. Bullone M, Lavoie JP. Asthma u201cof horses and menu201d-how can equine heaves help us better understand human asthma immunopathology and its functional consequences? Mol Immunol. 2015;66(1):97-105. https://doi.org/10.1016/j.molimm.2014.12.005
  75. Choy DF, Hart KM, Borthwick LA, Shikotra A, Nagarkar DR, Siddiqui S, et al. TH2 and TH17 inflammatory pathways are reciprocally regulated in asthma. Sci Transl Med. 2015;7(301):301ra129. https://doi.org/10.1126/scitranslmed.aab3142
  76. Davis KU, Sheats MK. The role of neutrophils in the pathophysiology of asthma in humans and horses. Inflammation. 2021;44(2):450-465. https://doi.org/10.1007/s10753-020-01362-2
  77. Janssen P, Tosi I, Hego A, Maru00e9chal P, Marichal T, Radermecker C. Neutrophil extracellular traps are found in bronchoalveolar lavage fluids of horses with severe asthma and correlate with asthma severity. Front Immunol. 2022;13:1-18. https://doi.org/10.3389/fimmu.2022.921077
  78. Burel JG, Pomaznoy M, Lindestam Arlehamn CS, Seumois G, Vijayanand P, Sette A, et al. The challenge of distinguishing cell-cell complexes from singlet cells in non-imaging flow cytometry and single-cell sorting. Cytom Part A. 2020;97(11):1127-1135. https://doi.org/10.1002/cyto.a.24027

Citations

This article has been cited 0 times.