C-Kit+ cells can modulate asthmatic condition via differentiation into pneumocyte-like cells and alteration of inflammatory responses via ERK/NF-ƙB pathway

Document Type : Original Article

Authors

1 Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran

2 Department of Physiology, Ardabil Branch, Islamic Azad University, Ardabil, Iran

3 Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran

4 Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran

5 Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran

6 Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran

7 Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran

Abstract

Objective(s): The exact role of the progenitor cell types in the dynamic healing of asthmatic lungs is lacking. This investigation was proposed to evaluate the effect of intratracheally administered rat bone marrow-derived c-kit+ cells on ovalbumin-induced sensitized male rats.
Materials and Methods: Forty rats were randomly divided into 4 groups; healthy rats received phosphate-buffered saline (PBS) (C); sensitized rats received PBS (S); PBS containing C-kitˉ cells (S+C-kit-); and PBS containing C-kit+ cells (S+C-kit+). After two weeks, circulatory CD4+/CD8+ T-cell counts and pulmonary ERK/NF-ƙB signaling pathway as well as the probability of cellular differentiation were assessed. 
Results: The results showed that transplanted C-Kit+ cells were engrafted into pulmonary tissue and differentiated into epithelial cells. C-Kit+ cells could increase the number of CD4+ cells in comparison with the S group (P<0.001); however, they diminished the level of CD8+ cells (P<0.01). Moreover, data demonstrated increased p-ERK/ERK ratio (P<0.001) and NF-ƙB level (P<0.05) in sensitized rats compared with the C group. The administration of C-kit+, but not C-Kit-, decreased p-ERK/ERK ratio and NF-ƙB level compared with those of the S group (P<0.05). 
Conclusion: The study revealed that C-Kit+ cells engrafted into pulmonary tissue reduced the NF-ƙB protein level and diminished p-ERK/ERK ratio, leading to suppression of inflammatory response in asthmatic lungs. 

Keywords


1. Pawankar R. Allergic diseases and asthma: A global public health concern and a call to action. World Allergy Organ J 2014; 7:1939-4551.
2. Papi A, Brightling C, Pedersen SE, Reddel HK. Asthma. Lancet 2018; 391:783-800.
3. Price MM, Oskeritzian CA, Falanga YT, Harikumar KB, Allegood JC, Alvarez SE, et al. A specific sphingosine kinase 1 inhibitor attenuates airway hyperresponsiveness and inflammation in a mast cell-dependent murine model of allergic asthma. J Allergy Clin Immunol 2013; 131:501-511.
4. Endo Y, Nakayama T. Pathogenic Th2 (Tpath2) cells in airway inflammation. Oncotarget 2015; 6:32303-32304.
5. Fahy JV. Type 2 inflammation in asthma--present in most, absent in many. Nat Rev Immunol 2015; 15:57-65.
6. Foster PS, Maltby S, Rosenberg HF, Tay HL, Hogan SP, Collison AM, et al. Modeling TH2 responses and airway inflammation to understand fundamental mechanisms regulating the pathogenesis of asthma. Immunol Rev 2017; 278:20-40.
7. Rogala B, Bozek A, Gluck J, Jarzab J. Prevalence of IgE-mediated allergy and evaluation of Th1/Th2 cytokine profiles in patients with severe bronchial asthma. Postepy Dermatol Alergol 2015; 32:274.
8. Guo C-H, Liu P-J, Hsia S, Chuang C-J, Chen P-C. Role of certain trace minerals in oxidative stress, inflammation, CD4/CD8 lymphocyte ratios and lung function in asthmatic patients. Ann Clin Biochem 2011; 48:344-351.
9. Miyahara N, Swanson BJ, Takeda K, Taube C, Miyahara S, Kodama T, et al. Effector CD8+ T cells mediate inflammation and airway hyper-responsiveness. Nat med 2004; 10:865-869.
10. Ci X, Chu X, Xu X, Li H, Deng X. Short-term roxithromycin treatment attenuates airway inflammation via MAPK/NF-κB activation in a mouse model of allergic asthma. Inflamm Res 2012; 61:749-758.
11. Zhang Y, Cardell L-O, Edvinsson L, Xu C-B. MAPK/NF-κB-dependent upregulation of kinin receptors mediates airway hyperreactivity: a new perspective for the treatment. Pharmacol Res 2013; 71:9-18.
12. Gras D, Chanez P, Vachier I, Petit A, Bourdin A. Bronchial epithelium as a target for innovative treatments in asthma. Pharmacol Ther 2013; 140:290-305.
13. Mishra A, Yao X, Levine SJ. From bedside to bench to clinic trials: identifying new treatments for severe asthma. Dis Model Mech 2013; 6:877-888.
14. Wise J. Corticosteroids for asthma may suppress growth in children in first year of treatment, researchers say. BMJ 2014; 349:g4623.
15. Ciriaco M, Ventrice P, Russo G, Scicchitano M, Mazzitello G, Scicchitano F, et al. Corticosteroid-related central nervous system side effects. J Pharmacol Pharmacother 2013; 4:S94.
16. Ogliari KS, Marinowic D, Brum DE, Loth F. Stem cells in dermatology. An bras dermatol 2014; 89:286-291.
17. Bogers SH. Cell-based therapies for joint disease in veterinary medicine: what we have learned and what we need to know. Front Vet Sci 2018; 5:70-86.
18. Arnhold S, Elashry MI, Klymiuk MC, Wenisch S. Biological macromolecules and mesenchymal stem cells: Basic research for regenerative therapies in veterinary medicine. Int J Biol Macromol 2019; 123:889.
19. Dias IE, Pinto PO, Barros LC, Viegas CA, Dias IR, Carvalho PP. Mesenchymal stem cells therapy in companion animals: useful for immune-mediated diseases? BMC Vet Res 2019; 15:358.
20. Marino F, Scalise M, Cianflone E, Mancuso T, Aquila I, Agosti V, et al. Role of c-Kit in myocardial regeneration and aging. Front Endocrinol 2019; 10.
21. Micheva-Viteva SN, Shou Y, Nowak-Lovato KL, Rector KD, Hong-Geller E. c-KIT signaling is targeted by pathogenic Yersinia to suppress the host immune response. BMC Microbiol 2013; 13:249.
22. Corti S, Nizzardo M, Nardini M, Donadoni C, Salani S, Simone C, et al. Systemic transplantation of c-kit+ cells exerts a therapeutic effect in a model of amyotrophic lateral sclerosis. Hum Mol Genet 2010; 19:3782-3796.
23. Moorefield EC, McKee EE, Solchaga L, Orlando G, Yoo JJ, Walker S, et al. Cloned, CD117 selected human amniotic fluid stem cells are capable of modulating the immune response. PLoS One 2011; 6-12.
24. Di Trapani M, Bassi G, Fontana E, Giacomello L, Pozzobon M, Guillot PV, et al. Immune regulatory properties of CD117pos amniotic fluid stem cells vary according to gestational age. Stem Cells Dev 2015; 24:132-143.
25. Zou T, Gao L, Zeng Y, Li Q, Li Y, Chen S, et al. Organoid-derived C-Kit+/SSEA4− human retinal progenitor cells promote a protective retinal microenvironment during transplantation in rodents. Nat commun 2019; 10:1-17.
26. Ellison GM, Vicinanza C, Smith AJ, Aquila I, Leone A, Waring CD, et al. Adult c-kitpos cardiac stem cells are necessary and sufficient for functional cardiac regeneration and repair. Cell 2013; 154:827-842.
27. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell 2003; 114:763-776.
28. Smith AJ, Lewis FC, Aquila I, Waring CD, Nocera A, Agosti V, et al. Isolation and characterization of resident endogenous c-Kit+ cardiac stem cells from the adult mouse and rat heart. Nat Protoc 2014; 9:1662-1681.
29. Ghobadi H, Alipour MR, Keyhanmanesh R, Boskabady MH, Aslani MR. Effect of high-fat diet on tracheal responsiveness to methacholine and insulin resistance index in ovalbumin-sensitized male and female rats. Iran J Allergy Asthma Immunol 2019; 18:48-61.
30. Akhavanakbari G, Babapour B, Alipour MR, Keyhanmanesh R, Ahmadi M, Aslani MR. Effect of high fat diet on NF-кB microRNA146a negative feedback loop in ovalbumin-sensitized rats. BioFactors 2019; 45:75-84.
31. Khaksar M, Sayyari M, Rezaie J, Pouyafar A, Montazersaheb S, Rahbarghazi R. High glucose condition limited the angiogenic/cardiogenic capacity of murine cardiac progenitor cells in in vitro and in vivo milieu. Cell Biochem Func 2018; 36:346-356.
32. Rahbarghazi R, Keyhanmanesh R, Aslani MR, Hassanpour M, Ahmadi M. Bone marrow mesenchymal stem cells and condition media diminish inflammatory adhesion molecules of pulmonary endothelial cells in an ovalbumin-induced asthmatic rat model. Microvasc Res 2019; 121:63-70.
33. Dong QM, Ling C, Zhao L. Immunofluorescence analysis of cytokeratin 8/18 staining is a sensitive assay for the detection of cell apoptosis. Oncol Lett 2015; 9:1227-1230.
34. Lindsey J, Ganguly K, Brass D, Li Z, Potts E, Degan S, et al. c-Kit is essential for alveolar maintenance and protection from emphysema-like disease in mice. Am J Respir Crit Care Med 2011; 183:1644–1652.
35. Spaziano G, Cappetta D, Urbanek K, Piegari E, Esposito G, Matteis M, et al. New role of adult lung c-kit+ cells in a mouse model of airway hyperresponsiveness. Mediators Inflamm 2016; 2016.
36. Kajstura J, Rota M, Hall SR, Hosoda T, D’Amario D, Sanada F, et al. Evidence for human lung stem cells. N Engl J Med 2011; 364:1795-1806.
37. Sen N, Weprin S, Peter Y. Discrimination between lung homeostatic and injury-induced epithelial progenitor subsets by cell-density properties. Stem Cells Dev 2013; 22:2036-2046.
38. Suzuki T, Suzuki S, Fujino N, Ota C, Yamada M, Suzuki T, et al. c-Kit immunoexpression delineates a putative endothelial progenitor cell population in developing human lungs. Am J Physiol-Lung Cell Mol Physiol 2014; 306:L855-L865.
39. Liu Q, Huang X, Zhang H, Tian X, He L, Yang R, et al. c-Kit+ cells adopt vascular endothelial but not epithelial cell fates during lung maintenance and repair. Nat Med 2015; 21:866-868.
40. Takahashi T, Friedmacher F, Zimmer J, Puri P. Increased c-kit and stem cell factor expression in the pulmonary vasculature of nitrofen-induced congenital diaphragmatic hernia. J Pediatric Surgery 2016; 51:706-709.
41. Pelaia G, Vatrella A, Busceti M, Gallelli L, Calabrese C, Terracciano R, et al. Cellular mechanisms underlying eosinophilic and neutrophilic airway inflammation in asthma. Mediators Inflamm 2015; 2015:1-8.
42. Ahmadi M, Rahbarghazi R, Soltani S, Aslani MR, Keyhanmanesh R. Contributory anti-inflammatory effects of mesenchymal stem cells, not conditioned media, on ovalbumin-induced asthmatic changes in male rats. Inflammation 2016; 39:1960-1971.
43. Ahmadi M, Rahbarghazi R, Aslani MR, Shahbazfar A-A, Kazemi M, Keyhanmanesh R. Bone marrow mesenchymal stem cells and their conditioned media could potentially ameliorate ovalbumin-induced asthmatic changes. Biomed Pharmacother 2017; 85:28-40.
44. Li P, Li Z, Zhang G, Yang J, Chen J. CD4+ CD25+ Regulatory T Cells Decreased CD8+ IL-4+ cellsin a mouse model of allergic asthma. Iranian J Allergy Asthma Immunol 2018:1-10.
45. Heiran H, Ahmadi M, Rahbarghazi R, Mir-ershadi F, Delkhosh A, Khaksar M, et al. C-Kit+ progenitors restore rat asthmatic lung function by modulation of T-bet and GATA-3 expression. Exp Physiol 2020; 105:1623-1633.
46. Rahbarghazi R, Keyhanmanesh R, Rezaie J, Mirershadi F, Heiran H, Saghaei Bagheri H, et al. c-Kit+ cells offer hopes in ameliorating asthmatic pathologies via regulation of miRNA-133 and -126. Iran J Basic Med Sci 2021; 24:1-8.
47. Mirershadi F, Ahmadi M, Rezabakhsh A, Rajabi H, Rahbarghazi R, Keyhanmanesh R. Unraveling the therapeutic effects of mesenchymal stem cells in asthma. Stem Cell Res Ther 2020; 11:400-411.
48. Damera G, Druey KM, Cooper PR, Krymskaya VP, Soberman RJ, Amrani Y, et al. An rgs4-mediated phenotypic switch of bronchial smooth muscle cells promotes fixed airway obstruction in asthma. PLoS ONE 2012; 7:e28504.
49. Schuliga M. NF-kappaB Signaling in chronic inflammatory airway disease. Biomolecules 2015; 5:1266-1283.
50. Shimizu K, Konno S, Ozaki M, Umezawa K, Yamashita K, Todo S, et al. Dehydroxymethylepoxyquinomicin (DHMEQ), a novel NF-kappaB inhibitor, inhibits allergic inflammation and airway remodelling in murine models of asthma. Clin Exp Allergy 2012; 42:1273–1281.
51. Moore F, Buonocore S, Aksoy E, Ouled-Haddou N, Goriely S, Lazarova E, et al. An alternative pathway of NF-kappaB activation results in maturation and T cell priming activity of dendritic cells overexpressing a mutated IkappaBalpha. J Immunol 2007; 178:1301-1311.
52. Hirose K, Wakashin H, Oki m, Kagami S, Suto A, Ikeda K, et al. GS143, an IkappaB ubiquitination inhibitor, inhibits allergic airway inflammation in mice. Biochem biophys Res Commun 2008; 374:507-511.
53. Piao H, Choi YH, Li H, Wang C, Xian Z, Ogasawara M, et al. Recombinant pyrin domain protein attenuates allergic inflammation by suppressing NF-κB pathway in asthmatic mice. Scandinavian J Immunol 2019; 89:e12720.
54. Kandhare AD, Liu Z, Mukherjee AA, Bodhankar SL. Therapeutic potential of morin in ovalbumin-induced allergic asthma via modulation of SUMF2/IL-13 and BLT2/NF-kB signaling pathway. Current Mol Pharmacol 2019; 12:122-138.
55. Pang L, Zou S, Shi Y, Mao Q, Chen Y. Apigenin attenuates PM2. 5-induced airway hyperresponsiveness and inflammation by down-regulating NF-κB in murine model of asthma. Int J Clin Exp Pathol 2019; 12:3700-3709.
56. Shin N-R, Lee AY, Song J-H, Yang S, Park I, Lim J-O, et al. Scrophularia buergeriana attenuates allergic inflammation by reducing NF-κB activation. Phytomedicine 2020; 67:153159.
57. Liu W, Liang Q, Balzar S, Wenzel S, Gorska M, Alam R. Cell-specific activation profile of extracellular signal-regulated kinase 1/2, jun n-terminal kinase, and p38 mitogen-activated protein kinases in asthmatic airways. J Allergy Clin Immunol 2008; 121:893-902 e892.
58. Gaffey K, Reynolds S, Plumb J, Kaur M, Singh D. Increased phosphorylated p38 mitogen activated protein kinase in copd lungs. Eur Respir J 2013; 42:28-41.
59. Munoz L, Ramsay E, Manetsch M, Ge Q, Peifer C, Laufer S, et al. Novel p38 MAPK inhibitor ml3403 has potent anti-inflammatory activity in airway smooth muscle. Eur J Pharmacol 2010; 635:212–218.
60. Chung K. P38 mitogen-activated protein kinase pathways in asthma and COPD. Chest 2011; 139:1470–1479.