• 沒有找到結果。

Figure 1. Identification of neonatal PSCs. (A) Neonatal pulmonary cells were labeled

with Sca-1 and SSEA-1 antibodies and analyzed using flow cytometry. The data are representative of five independent experiments. (B) Representative flow cytometry histograms of SSEA-1 and Sca-1-stained cell population in the lung single-cell suspension. (C) Cell number of SSEA-1+- and Sca-1+- enriched pulmonary cell extracted from whole lung/mouse (n = 3-5 mice per group). Data are means ± SD and represent three independent experiments. ** P <0.01.

Figure 2. Whole-mount view of SSEA-1

+

pulmonary cells in lung tissues. Neonatal

and adult mice derived lung sections were stained with anti-SSEA-1 (green) and the nuclei were counterstained with DAPI (blue). Bar: 500 µm. Data are representative of at least five independent experiments.

Figure 3. Expression of SSEA-1

+

pulmonary cells in lung tissues. Neonatal and adult

mice derived lung sections were stained with anti-SSEA-1 (green) and CCSP (red); the nuclei were counterstained with DAPI (blue). The bronchiole (Br) and terminal bronchiolar (TB) airway were determined by CCSP (red) staining. Neonatal (left) and adult (right) SSEA-1+ pulmonary cells were located within TB adjacent to the bronchioalveolar duct junction (BADJ). Bars: 50 µm. The arrows point to the BADJ.

Data are representative of at least five independent experiments.

Figure 4. Characterization of neonatal SSEA-1

+

pulmonary cells. (A) Flow

cytometric analysis of surface marker expression in neonatal SSEA-1+ pulmonary cells.

The gray areas represent matched isotype controls. Data are representative of three independent experiments. (B) FACS analysis of intracellular SPC and surface T1α expression in SSEA-1+ PSCs. SSEA-1+ PSCs expressed SPC (a type II pneumocyte marker) but were negative for T1α (a type I pneumocyte marker). Data are representative of two independent experiments. (C) Enriched SSEA-1+ PSCs were purified from neonatal mice, and the CCSP expression levels were determined using RT-QPCR with specific primers. The mRNA levels were normalized using the housekeeping gene GAPDH. Bars indicate mean ± SD of three independent

prepared, and the CCSP protein levels were determined by immunoblots stained with a specific antibody against CCSP. β -actin was used as an internal control. Data are representative of two independent experiments.

Figure 5. Neonatal SSEA-1

+

pulmonary cells are negative for pneumocyte-related

markers. Newborn mice-derived pulmonary single-cell suspensions and paraffin-fixed

lung tissues were stained with SSEA-1 (green), alveolar cell markers (red) AQP5 (a type I pneumocyte marker), and proSPC (a type II pneumocyte marker). The nuclei were counterstained with DAPI (blue). Bars, 20 µm.

Figure 6. Neonatal SSEA-1

+

pulmonary cells are self-renewal and clonogenicity. (A)

Representative photographs of sphere colonies cultured in Matrigel with Transwell plates. (B) Representative photographs of primary and secondary sphere colonies. Bars, 100 µm. (C) Efficiency of secondary spheres formation. The results are expressed as means ± SD (n = 3).

Figure 7. Neonatal SSEA-1

+

pulmonary cells are multipotent. (A) SSEA-1

+ PSCs cultured on Matrigel-coated plate for 15-20 days were stained with pro SPC (green; left) and AQP5 (green; right) antibody to identify the type I pneumocyte; the nuclei were counterstained with DAPI (blue). Bars, 20 µm. (B) SSEA-1+ PSCs seeded in an ALI culture for 15 days were labeled for ZO-1 (green), γ -tubulin (red), and DAPI (blue).

Bars, 20 µm. (C) Scanning electron microscopic images of in vitro-cultured SSEA-1+ PSCs at ALI for 15 days, showing ciliated and nonciliated cells. Bar, 10 µm.

Figure 8. Neonatal SSEA-1

+

PSCs suppress TSLP and eotaxin production. Lung

epithelial cells (2×104) derived from adult mice were co-cultured with neonatal SSEA-1+ PSCs (1×104) in the presence of (A) LPS (2 µg/ml) or (B) IL-4 (100 ng/ml) for 24 hours. TSLP and eotaxin contents in culture supernatants were determined using ELISA.

Values are the means ± SD. The data are representative of three independent experiments. * P < 0.05, ** P < 0.01.

Figure 9. SSEA-1

+

PSCs suppress TSLP and eotaxin production through cell-cell contact independent manner, respectively. Co-culture system of adult lung epithelial

cells (2×104; bottom) and neonatal SSEA-1+ PSCs (1×104; insert) were separated with a Tranwell and stimulated with (A) LPS (2 µg/ml) or (B) IL-4 (100 ng/ml) for 24 hours.

TSLP and eotaxin contents in culture supernatants were determined using ELISA.

Values are the means ± SD. The data are representative of three independent experiments. * P < 0.05, ** P < 0.01.

Figure 10. Neonatal lung SSEA-1

+

PSCs reduced TSLP and eotaxin production from adult lung epithelial cells through CCSP-dependent and -independent manner. Lung epithelial cells derived from adult mice were co-cultured with neonatal

SSEA-1+ PSCs and stimulated with (A) LPS or (B) IL-4 for 24 hours in the presence of control antibody or CCSP neutralizing antibody (5 µg/ml). TSLP and eotaxin contents in LPS- and IL-4-stimulated culture supernatants were determined using ELISA, respectively. Data are means ± SD and representative of three independent experiments.

** P < 0.01.

Figure 11. Transplantation of SSEA-1

+

PSCs did not change serum OVA-specific

IgE in the asthmatic mice. (A) Distribution of SSEA-1

+ PSCs in adoptive transferred mice. The lung sections were stained with an anti-GFP- (green) and an anti-panCK (red) antibody, and they were counterstained with DAPI (blue). Bars, 50 µm. (B) Magnified view of the boxed area in A. Bar, 50 µm. (C) Transplanted PSCs maintained SSEA-1 expression after repeated OVA challenge in vivo. The lung sections were stained with an anti-GFP- (green) and an anti-SSEA-1 (red) antibody, and they were counterstained with DAPI (blue). Bar, 20 µm. (D) The mice received an i.v. injection of 1×105 cells of SSEA-1+ PSCs or SSEA-1- pulmonary cells isolated from neonatal GFP-mice after the second aerosol treatment. OVA-specific IgE was detected in serum 24 hours after the last inhalation exposure to OVA. Data are means ± SEM and representative of two independent experiments (n = 5 mice per group).

Figure 12. Transplantation of SSEA-1

+

PSCs before the OVA challenge reduced

the level of inflammatory mediators and the cellular infiltration of the lungs. 1×10

5 SSEA-1+ PSCs isolated from neonatal mice were i.v. injected into OVA-induced asthmatic mice prior to an OVA aerosol challenge. (A) The BAL fluid was taken 24 hours after the last aerosol OVA exposure. Differential counts of the cells in the BAL fluid were obtained by FACS analysis, and (B) The levels of IL-4, IL-5, TSLP, and eotaxin in the BAL fluid was determined by ELISA. n = 3-5 mice per group. Bars represent means ± SD. * P < 0.05, ** P <0.01. The data from one representative experiment of two are shown.

Figure 13. Transplantation of SSEA-1

+

PSCs improves AHR and airway

eosinophilia in the asthmatic mice. (A) Flowchart of the method used to produce the

OVA-induced asthmatic murine model. (B) Airway function was measured by invasive body plethysmography. The results are expressed as means ± SEM of the lung resistance (RL) in the ratio of RL after PBS nebulization. Data are represent two independent experiments (n = 4-7 mice per group). ** P < 0.01. (C) Cell number in the recovered BAL fluid was counted after staining by trypan blue. (D) Eosinophil counts in the BAL fluid were obtained by FACS analysis. Data are means ± SEM and representative of two independent experiments (n = 6-8 mice per group). ** P < 0.01.

Figure 14. Transplantation of SSEA-1

+

PSCs reduces airway inflammatory

cytokine/chemokine production in the asthmatic mice. BAL fluid was taken 24 hours

after the last aerosol OVA exposure. The level of eotaxin, TSLP, IL-4, IL-5, and IL-13 in BAL fluid was determined using specific ELISA kits. Data are means ± SEM and representative of two independent experiments (n = 4-6 mice per group). ** P < 0.01.

Figure. 15. Transplantation of SSEA-1

+

PSCs reduces airway inflammation. H&E

staining of lung sections in untreated, SSEA-1+ PSCs-, SSEA-1- pulmonary cells-treated, and healthy group. Bars, 200 µm. Arrows denote infiltrated leukocytes; br, bronchus; v, vessel. Data are representative of two independent experiments.

Figure 16. Transplantation of SSEA-1

+

PSCs decreased the infiltration of

inflammatory cells into lung in the asthmatic mice. Enlarge view of Figure 15. Bars,

200 µm. Arrows denote infiltrated leukocytes; br, bronchus; v, vessel.

Figure 17. Transplantation of SSEA-1

+

PSCs increase Foxp3

+

Treg population.

Total cell number in (A) thoracic and (B) cervical LN. Cell number and percentage of Foxp3-GFP+ CD4+ Treg in (C) thoracic and (D) cervical LN determined by FACS analysis (n = 5-9 mice/group). Values are presented as the means ± SD. The data are representative of two independent experiments. * P < 0.05, ** P <0.01.

Figure 18. Transplantation of SSEA-1

+

PSCs preserve CCSP production. (A) The

expression of CCSP in lung sections from untreated-, SSEA-1+ PSCs-treated-, and healthy mice was analyzed using immunostaining with an anti-CCSP antibody (green);

and the nuclei were counterstained with DAPI (blue). The data are representative of two independent experiments. Bars, 50 µm. (B) The transcript levels of CCSP in lung tissues were analyzed using RT-QPCR with specific primers. The mRNA levels were normalized with GAPDH as a housekeeping gene. * P < 0.05, ** P <0.01.

Figure 19. Transplantation of SSEA-1

+

PSCs inhibits unregulated cell proliferation in asthma. (A) Schematic representation of an OVA-induced asthma model with BrdU

administration. (B) Representative BrdU immunostaining of proliferating cells (red);

and the nuclei were counterstained with DAPI (blue). The data are representative of two independent experiments. Bars, 100 µm. (C) Graphic representation of decreased BrdU incorporation in the lungs of SSEA-1+ PSCs-recipient mice. Two to three consecutive cryosections from each mouse lung were examined, and the average number of BrdU-positive nuclei per 1,000 nuclei was used as the value for the mouse. The data are expressed as the means ± SD, n = 3 mice/group. ** P < 0.01.

Figure 20. BrdU-incorporated cell population in asthma. Immunofluorescence

staining of T1α, CD3 or TTF-1 (green), BrdU (red) and DAPI (blue) in asthmatic lung tissue. Bars, 20 µm.

VII. REFERENCES

1. Busse WW, Lemanske RF, Jr. Asthma. N Engl J Med 2001; 344:350-62.

2. Grainge CL, Davies DE. Epithelial injury and repair in airways diseases. Chest 2013; 144:1906-12.

3. Bousquet J, Kaltaev N. Global surveillance, prevention and control of chronic respiratory diseases : a comprehensive approach. Geneva: World Health Organization; 2007.

4. Dahl R. Systemic side effects of inhaled corticosteroids in patients with asthma.

Respir Med 2006; 100:1307-17.

5. Sueblinvong V, Weiss DJ. Stem cells and cell therapy approaches in lung biology and diseases. Transl Res 2010; 156:188-205.

6. Kim HY, DeKruyff RH, Umetsu DT. The many paths to asthma: phenotype shaped by innate and adaptive immunity. Nat Immunol 2010; 11:577-84.

7. Johnston RA, Zhu M, Rivera-Sanchez YM, Lu FL, Theman TA, Flynt L, et al.

Allergic airway responses in obese mice. Am J Respir Crit Care Med 2007;

176:650-8.

8. Pichavant M, Goya S, Meyer EH, Johnston RA, Kim HY, Matangkasombut P, et al. Ozone exposure in a mouse model induces airway hyperreactivity that

requires the presence of natural killer T cells and IL-17. J Exp Med 2008;

205:385-93.

9. Kim EY, Battaile JT, Patel AC, You Y, Agapov E, Grayson MH, et al.

Persistent activation of an innate immune response translates respiratory viral infection into chronic lung disease. Nat Med 2008; 14:633-40.

10. Wamboldt MZ, Laudenslager M, Wamboldt FS, Kelsay K, Hewitt J.

Adolescents with atopic disorders have an attenuated cortisol response to

11. Larche M, Akdis CA, Valenta R. Immunological mechanisms of allergen-specific immunotherapy. Nat Rev Immunol 2006; 6:761-71.

12. Palomares O, Yaman G, Azkur AK, Akkoc T, Akdis M, Akdis CA. Role of Treg in immune regulation of allergic diseases. Eur J Immunol 2010; 40:1232-40.

13. Lloyd CM, Hessel EM. Functions of T cells in asthma: more than just T(H)2 cells. Nat Rev Immunol 2010; 10:838-48.

14. Gould HJ, Sutton BJ. IgE in allergy and asthma today. Nat Rev Immunol 2008;

8:205-17.

15. Paul WE, Zhu J. How are T(H)2-type immune responses initiated and amplified?

Nat Rev Immunol 2010; 10:225-35.

16. Takatsu K, Nakajima H. IL-5 and eosinophilia. Curr Opin Immunol 2008;

20:288-94.

17. Rosenberg HF, Dyer KD, Foster PS. Eosinophils: changing perspectives in health and disease. Nat Rev Immunol 2013; 13:9-22.

18. Ingram JL, Kraft M. IL-13 in asthma and allergic disease: asthma phenotypes and targeted therapies. J Allergy Clin Immunol 2012; 130:829-42; quiz 43-4.

19. Wills-Karp M. Interleukin-13 in asthma pathogenesis. Immunol Rev 2004;

202:175-90.

20. Cohn L, Homer RJ, Marinov A, Rankin J, Bottomly K. Induction of airway mucus production By T helper 2 (Th2) cells: a critical role for interleukin 4 in cell recruitment but not mucus production. J Exp Med 1997; 186:1737-47.

21. Wing K, Onishi Y, Prieto-Martin P, Yamaguchi T, Miyara M, Fehervari Z, et al.

CTLA-4 control over Foxp3+ regulatory T cell function. Science 2008;

322:271-5.

22. Akdis M, Verhagen J, Taylor A, Karamloo F, Karagiannidis C, Crameri R, et al.

Immune responses in healthy and allergic individuals are characterized by a fine balance between allergen-specific T regulatory 1 and T helper 2 cells. J Exp Med 2004; 199:1567-75.

23. Gri G, Piconese S, Frossi B, Manfroi V, Merluzzi S, Tripodo C, et al.

CD4+CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40-OX40L interaction. Immunity 2008; 29:771-81.

24. Ring S, Schafer SC, Mahnke K, Lehr HA, Enk AH. CD4+ CD25+ regulatory T cells suppress contact hypersensitivity reactions by blocking influx of effector T cells into inflamed tissue. Eur J Immunol 2006; 36:2981-92.

25. Joetham A, Takeda K, Taube C, Miyahara N, Matsubara S, Koya T, et al.

Naturally occurring lung CD4(+)CD25(+) T cell regulation of airway allergic responses depends on IL-10 induction of TGF-beta. J Immunol 2007; 178:1433-42.

26. Kearley J, Barker JE, Robinson DS, Lloyd CM. Resolution of airway inflammation and hyperreactivity after in vivo transfer of CD4+CD25+

regulatory T cells is interleukin 10 dependent. J Exp Med 2005; 202:1539-47.

27. Ostroukhova M, Seguin-Devaux C, Oriss TB, Dixon-McCarthy B, Yang L, Ameredes BT, et al. Tolerance induced by inhaled antigen involves CD4(+) T cells expressing membrane-bound TGF-beta and FOXP3. J Clin Invest 2004;

114:28-38.

28. Strickland DH, Stumbles PA, Zosky GR, Subrata LS, Thomas JA, Turner DJ, et al. Reversal of airway hyperresponsiveness by induction of airway mucosal CD4+CD25+ regulatory T cells. J Exp Med 2006; 203:2649-60.

29. Nguyen KD, Vanichsarn C, Fohner A, Nadeau KC. Selective deregulation in chemokine signaling pathways of CD4+CD25(hi)CD127(lo)/(-) regulatory T cells in human allergic asthma. J Allergy Clin Immunol 2009; 123:933-9 e10.

30. Hartl D, Koller B, Mehlhorn AT, Reinhardt D, Nicolai T, Schendel DJ, et al.

Quantitative and functional impairment of pulmonary CD4+CD25hi regulatory T cells in pediatric asthma. J Allergy Clin Immunol 2007; 119:1258-66.

31. Fulkerson PC, Rothenberg ME. Targeting eosinophils in allergy, inflammation and beyond. Nat Rev Drug Discov 2013; 12:117-29.

32. Brottman GM, Regelmann WE, Slungaard A, Wangensteen OD. Effect of eosinophil peroxidase on airway epithelial permeability in the guinea pig.

Pediatr Pulmonol 1996; 21:159-66.

33. Gleich GJ, Frigas E, Loegering DA, Wassom DL, Steinmuller D. Cytotoxic properties of the eosinophil major basic protein. J Immunol 1979; 123:2925-7.

34. Kampen GT, Stafford S, Adachi T, Jinquan T, Quan S, Grant JA, et al. Eotaxin induces degranulation and chemotaxis of eosinophils through the activation of ERK2 and p38 mitogen-activated protein kinases. Blood 2000; 95:1911-7.

35. Tenscher K, Metzner B, Schopf E, Norgauer J, Czech W. Recombinant human eotaxin induces oxygen radical production, Ca(2+)-mobilization, actin

reorganization, and CD11b upregulation in human eosinophils via a pertussis toxin-sensitive heterotrimeric guanine nucleotide-binding protein. Blood 1996;

88:3195-9.

36. Ying S, Robinson DS, Meng Q, Rottman J, Kennedy R, Ringler DJ, et al.

Enhanced expression of eotaxin and CCR3 mRNA and protein in atopic asthma.

Association with airway hyperresponsiveness and predominant co-localization

of eotaxin mRNA to bronchial epithelial and endothelial cells. Eur J Immunol 1997; 27:3507-16.

37. Schuh JM, Blease K, Kunkel SL, Hogaboam CM. Eotaxin/CCL11 is involved in acute, but not chronic, allergic airway responses to Aspergillus fumigatus. Am J Physiol Lung Cell Mol Physiol 2002; 283:L198-204.

38. Pope SM, Zimmermann N, Stringer KF, Karow ML, Rothenberg ME. The eotaxin chemokines and CCR3 are fundamental regulators of allergen-induced pulmonary eosinophilia. J Immunol 2005; 175:5341-50.

39. Ma W, Bryce PJ, Humbles AA, Laouini D, Yalcindag A, Alenius H, et al. CCR3 is essential for skin eosinophilia and airway hyperresponsiveness in a murine model of allergic skin inflammation. J Clin Invest 2002; 109:621-8.

40. Fulkerson PC, Fischetti CA, McBride ML, Hassman LM, Hogan SP,

Rothenberg ME. A central regulatory role for eosinophils and the eotaxin/CCR3 axis in chronic experimental allergic airway inflammation. Proc Natl Acad Sci U S A 2006; 103:16418-23.

41. Kim J, Merry AC, Nemzek JA, Bolgos GL, Siddiqui J, Remick DG. Eotaxin represents the principal eosinophil chemoattractant in a novel murine asthma model induced by house dust containing cockroach allergens. J Immunol 2001;

167:2808-15.

42. Lambrecht BN, Hammad H. The airway epithelium in asthma. Nat Med 2012;

18:684-92.

43. Hammad H, Lambrecht BN. Dendritic cells and epithelial cells: linking innate and adaptive immunity in asthma. Nat Rev Immunol 2008; 8:193-204.

44. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN.

House dust mite allergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med 2009; 15:410-6.

45. Willart MA, Deswarte K, Pouliot P, Braun H, Beyaert R, Lambrecht BN, et al.

Interleukin-1alpha controls allergic sensitization to inhaled house dust mite via the epithelial release of GM-CSF and IL-33. J Exp Med 2012; 209:1505-17.

46. Licona-Limon P, Kim LK, Palm NW, Flavell RA. TH2, allergy and group 2 innate lymphoid cells. Nat Immunol 2013; 14:536-42.

47. Saenz SA, Taylor BC, Artis D. Welcome to the neighborhood: epithelial cell-derived cytokines license innate and adaptive immune responses at mucosal sites.

Immunol Rev 2008; 226:172-90.

48. Semlali A, Jacques E, Koussih L, Gounni AS, Chakir J. Thymic stromal lymphopoietin-induced human asthmatic airway epithelial cell proliferation through an IL-13-dependent pathway. J Allergy Clin Immunol 2010; 125:844-50.

49. Ito T, Wang YH, Duramad O, Hori T, Delespesse GJ, Watanabe N, et al. TSLP-activated dendritic cells induce an inflammatory T helper type 2 cell response through OX40 ligand. J Exp Med 2005; 202:1213-23.

50. Zhou B, Comeau MR, De Smedt T, Liggitt HD, Dahl ME, Lewis DB, et al.

Thymic stromal lymphopoietin as a key initiator of allergic airway inflammation in mice. Nat Immunol 2005; 6:1047-53.

51. He R, Oyoshi MK, Garibyan L, Kumar L, Ziegler SF, Geha RS. TSLP acts on infiltrating effector T cells to drive allergic skin inflammation. Proc Natl Acad Sci U S A 2008; 105:11875-80.

52. Boehme SA, Franz-Bacon K, Chen EP, Sasik R, Sprague LJ, Ly TW, et al. A small molecule CRTH2 antagonist inhibits FITC-induced allergic cutaneous inflammation. Int Immunol 2009; 21:81-93.

53. Al-Shami A, Spolski R, Kelly J, Keane-Myers A, Leonard WJ. A role for TSLP in the development of inflammation in an asthma model. J Exp Med 2005;

202:829-39.

54. Asquith KL, Ramshaw HS, Hansbro PM, Beagley KW, Lopez AF, Foster PS.

The IL-3/IL-5/GM-CSF common receptor plays a pivotal role in the regulation of Th2 immunity and allergic airway inflammation. J Immunol 2008; 180:1199-206.

55. Stampfli MR, Wiley RE, Neigh GS, Gajewska BU, Lei XF, Snider DP, et al.

GM-CSF transgene expression in the airway allows aerosolized ovalbumin to induce allergic sensitization in mice. J Clin Invest 1998; 102:1704-14.

56. Cates EC, Fattouh R, Wattie J, Inman MD, Goncharova S, Coyle AJ, et al.

Intranasal exposure of mice to house dust mite elicits allergic airway

inflammation via a GM-CSF-mediated mechanism. J Immunol 2004; 173:6384-92.

57. Bleck B, Tse DB, Jaspers I, Curotto de Lafaille MA, Reibman J. Diesel exhaust particle-exposed human bronchial epithelial cells induce dendritic cell

maturation. J Immunol 2006; 176:7431-7.

58. Ohta K, Yamashita N, Tajima M, Miyasaka T, Nakano J, Nakajima M, et al.

Diesel exhaust particulate induces airway hyperresponsiveness in a murine model: essential role of GM-CSF. J Allergy Clin Immunol 1999; 104:1024-30.

59. Schmitz N, Kurrer M, Kopf M. The IL-1 receptor 1 is critical for Th2 cell type airway immune responses in a mild but not in a more severe asthma model. Eur J Immunol 2003; 33:991-1000.

60. Eiwegger T, Akdis CA. IL-33 links tissue cells, dendritic cells and Th2 cell development in a mouse model of asthma. Eur J Immunol 2011; 41:1535-8.

61. Kurowska-Stolarska M, Stolarski B, Kewin P, Murphy G, Corrigan CJ, Ying S, et al. IL-33 amplifies the polarization of alternatively activated macrophages that contribute to airway inflammation. J Immunol 2009; 183:6469-77.

62. Chu DK, Llop-Guevara A, Walker TD, Flader K, Goncharova S, Boudreau JE, et al. IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization. J Allergy Clin Immunol 2013; 131:187-200 e1-8.

63. Lambrecht BN, De Veerman M, Coyle AJ, Gutierrez-Ramos JC, Thielemans K, Pauwels RA. Myeloid dendritic cells induce Th2 responses to inhaled antigen, leading to eosinophilic airway inflammation. J Clin Invest 2000; 106:551-9.

64. Wills-Karp M, Rani R, Dienger K, Lewkowich I, Fox JG, Perkins C, et al.

Trefoil factor 2 rapidly induces interleukin 33 to promote type 2 immunity during allergic asthma and hookworm infection. J Exp Med 2012; 209:607-22.

65. Petersen BC, Budelsky AL, Baptist AP, Schaller MA, Lukacs NW. Interleukin-25 induces type 2 cytokine production in a steroid-resistant interleukin-17RB+

myeloid population that exacerbates asthmatic pathology. Nat Med 2012;

18:751-8.

66. Gregory LG, Jones CP, Walker SA, Sawant D, Gowers KH, Campbell GA, et al.

IL-25 drives remodelling in allergic airways disease induced by house dust mite.

67. Herriges M, Morrisey EE. Lung development: orchestrating the generation and regeneration of a complex organ. Development 2014; 141:502-13.

68. Hogan BL, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia CC, et al.

Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell Stem Cell 2014; 15:123-38.

69. Rawlins EL, Clark CP, Xue Y, Hogan BL. The Id2+ distal tip lung epithelium contains individual multipotent embryonic progenitor cells. Development 2009;

136:3741-5.

70. McQualter JL, Yuen K, Williams B, Bertoncello I. Evidence of an epithelial stem/progenitor cell hierarchy in the adult mouse lung. Proc Natl Acad Sci U S A 2010; 107:1414-9.

71. Rawlins EL. Lung epithelial progenitor cells: lessons from development. Proc Am Thorac Soc 2008; 5:675-81.

72. Kotton DN, Morrisey EE. Lung regeneration: mechanisms, applications and emerging stem cell populations. Nat Med 2014; 20:822-32.

73. Paul MK, Bisht B, Darmawan DO, Chiou R, Ha VL, Wallace WD, et al.

Dynamic changes in intracellular ROS levels regulate airway basal stem cell homeostasis through Nrf2-dependent Notch signaling. Cell Stem Cell 2014;

15:199-214.

74. Rock JR, Onaitis MW, Rawlins EL, Lu Y, Clark CP, Xue Y, et al. Basal cells as stem cells of the mouse trachea and human airway epithelium. Proc Natl Acad Sci U S A 2009; 106:12771-5.

75. Tata PR, Mou H, Pardo-Saganta A, Zhao R, Prabhu M, Law BM, et al.

Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature

76. Wansleeben C, Bowie E, Hotten DF, Yu YR, Hogan BL. Age-related changes in the cellular composition and epithelial organization of the mouse trachea. PLoS One 2014; 9:e93496.

77. Rock JR, Gao X, Xue Y, Randell SH, Kong YY, Hogan BL. Notch-dependent differentiation of adult airway basal stem cells. Cell Stem Cell 2011; 8:639-48.

78. Chen G, Korfhagen TR, Xu Y, Kitzmiller J, Wert SE, Maeda Y, et al. SPDEF is required for mouse pulmonary goblet cell differentiation and regulates a network of genes associated with mucus production. J Clin Invest 2009; 119:2914-24.

79. Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR, Stripp BR, et al.

Type 2 alveolar cells are stem cells in adult lung. J Clin Invest 2013; 123:3025-36.

80. Xu X, Rock JR, Lu Y, Futtner C, Schwab B, Guinney J, et al. Evidence for type II cells as cells of origin of K-Ras-induced distal lung adenocarcinoma. Proc Natl Acad Sci U S A 2012; 109:4910-5.

81. Desai TJ, Brownfield DG, Krasnow MA. Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature 2014; 507:190-4.

82. Reynolds SD, Reynolds PR, Pryhuber GS, Finder JD, Stripp BR. Secretoglobins SCGB3A1 and SCGB3A2 define secretory cell subsets in mouse and human airways. Am J Respir Crit Care Med 2002; 166:1498-509.

83. Giangreco A, Reynolds SD, Stripp BR. Terminal bronchioles harbor a unique airway stem cell population that localizes to the bronchoalveolar duct junction.

Am J Pathol 2002; 161:173-82.

84. Reynolds SD, Hong KU, Giangreco A, Mango GW, Guron C, Morimoto Y, et al.

Conditional clara cell ablation reveals a self-renewing progenitor function of

pulmonary neuroendocrine cells. Am J Physiol Lung Cell Mol Physiol 2000;

278:L1256-63.

85. Rawlins EL, Okubo T, Xue Y, Brass DM, Auten RL, Hasegawa H, et al. The role of Scgb1a1+ Clara cells in the long-term maintenance and repair of lung airway, but not alveolar, epithelium. Cell Stem Cell 2009; 4:525-34.

86. Johansson S, Wennergren G, Aberg N, Rudin A. Clara cell 16-kd protein downregulates T(H)2 differentiation of human naive neonatal T cells. J Allergy Clin Immunol 2007; 120:308-14.

86. Johansson S, Wennergren G, Aberg N, Rudin A. Clara cell 16-kd protein downregulates T(H)2 differentiation of human naive neonatal T cells. J Allergy Clin Immunol 2007; 120:308-14.

相關文件