Timing of umbilical cord blood derived mesenchymal stem cells transplantation determines therapeutic efficacy in the neonatal hyperoxic lung injury

Yun Sil Chang, Soo Jin Choi, So Yoon Ahn, Dong Kyung Sung, Se In Sung, Hye Soo Yoo, Won Il Oh, Won Soon Park, Yun Sil Chang, Soo Jin Choi, So Yoon Ahn, Dong Kyung Sung, Se In Sung, Hye Soo Yoo, Won Il Oh, Won Soon Park

Abstract

Intratracheal transplantation of human umbilical cord blood (UCB)-derived mesenchymal stem cells (MSCs) attenuates the hyperoxia-induced neonatal lung injury. The aim of this study was to optimize the timing of MSCs transplantation. Newborn Sprague-Dawley rats were randomly exposed to hyperoxia (90% for 2 weeks and 60% for 1 week) or normoxia after birth for 21 days. Human UCB-derived MSCs (5×10(5) cells) were delivered intratracheally early at postnatal day (P) 3 (HT3), late at P10 (HT10) or combined early+late at P3+10 (HT3+10). Hyperoxia-induced increase in mortality, TUNEL positive cells, ED1 positive alveolar macrophages, myeloperoxidase activity and collagen levels, retarded growth and reduced alveolarization as evidenced by increased mean linear intercept and mean alveolar volume were significantly better attenuated in both HT3 and HT3+10 than in HT10. Hyperoxia-induced up-regulation of both cytosolic and membrane p47(phox) indicative of oxidative stress, and increased inflammatory markers such as tumor necrosis factor-α, interleukin (IL) -1α, IL-1β, IL-6, and transforming growth factor-β measured by ELISA, and tissue inhibitor of metalloproteinase-1, CXCL7, RANTES, L-selectin and soluble intercellular adhesion molecule-1 measured by protein array were consistently more attenuated in both HT3 and HT3+10 than in HT10. Hyperoxia-induced decrease in hepatocyte growth factor and vascular endothelial growth factor was significantly up-regulated in both HT3 and HT3+10, but not in HT10. In summary, intratracheal transplantation of human UCB derived MSCs time-dependently attenuated hyperoxia-induced lung injury in neonatal rats, showing significant protection only in the early but not in the late phase of inflammation. There were no synergies with combined early+late MSCs transplantation.

Conflict of interest statement

Competing Interests: The authors have the following interests: co-authors Soo Jin Choi and Won-il Oh are employed by MEDIPOST Co., Ltd. Co-author Won-il Oh is a board member and holds stocks in MEDIPOST Co., Ltd. Samsung Medical Center and MEDIPOST Co., Ltd. have issued or filed patents for “Method of treating lung diseases using cells separated or proliferated from umbilical cord blood” under the name of Yun Sil Chang, Won Soon Park and Yoon Sun Yang. The relevent application number is: PCT/KR2007/000535. There are no further patents, products in development or marketed products to declare. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Temporal profiles of inflammatory cytokines.
Figure 1. Temporal profiles of inflammatory cytokines.
Tumor necrosis factor (TNF)-α, interleukin (IL)-1α, IL-1β, and IL-6 levels measured with ELISA at P 1, 3, 5, 7, 10 and 14 in the rat lung tissue. NC, Normoxia control group; HC, hyperoxia control group. Data; mean±SEM. *P<0.05 compared to NC.
Figure 2. Survival curve.
Figure 2. Survival curve.
Kaplan-Meier survival curve up to P 21 showing decreased survival. HC compared to NC, and improved survival in both HT3 and HT3+10, but not in HT10. NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. *P<0.05 compared to NC.
Figure 3. Histology and morphometric analysis of…
Figure 3. Histology and morphometric analysis of the surviving P21 rat lung.
(A): Representative optical microscopy photomicrographs of the lungs stained with hematoxylin and eosin (scale bar = 100 µm). (B): Degree of alveolarization measured by the mean linear intercept (left) and mean alveolar volume (right). NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. Data; mean±SEM. *P<0.05 compared to NC, #P<0.05 compared to HC,†P<0.05 compared to HT3, ‡P<0.05 compared to HT10.
Figure 4. TUNEL positive cells in the…
Figure 4. TUNEL positive cells in the distal lungs of the P 21 rat pups.
(A): TUNEL positive cells were labeled with FITC (green) and the cell nuclei were labeled with propidium iodide (red) (Scale bar; 25 µm). (B): Number of observed TUNEL positive cells per high power field. NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. Data; mean±SEM. *P<0.05 compared to NC, #P<0.05 compared to HC,†P<0.05 compared to HT3, ‡P<0.05 compared to HT10.
Figure 5. Donor cell localization in the…
Figure 5. Donor cell localization in the lung of the P 21 rats.
(A): Fluorescent microscopic observation of the PKH26 labeled human UCB-derived MSCs (donor cells, red) localized in the lungs of the P 21 newborn rats and the nuclei labeled with DAPI (blue) (Scale bar; 25 µm). (B): Number of PKH26 positive cells in the lung per high power field. NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. Data; mean±SEM. †P<0.05 compared to HT3.
Figure 6. p47 phox , cytosolic subunit…
Figure 6. p47phox, cytosolic subunit of nicotinamide adenine dinucleotide phosphate oxidase in the P21 rat lung tissue.
(A): Fluorescent microscopic observation of p47phox (green) localized in the lungs of the P21 rats and the nuclei labeled with DAPI (blue) (Scale bar; 25 µm). (B): Representative western blots (top) and densitometric histograms (bottom) in the cytosol (left) and membrane (right) fractions of P21 rat lung homogenates. NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. Data; mean±SEM. *P<0.05 compared to NC, #P<0.05 compared to HC.
Figure 7. Histograms of inflammatory cytokines and…
Figure 7. Histograms of inflammatory cytokines and chemokines in the hyperoxic lung injury after MSCs transplantation.
TNF-α, IL-1α, IL-1β, IL-6, and TGF-β levels measured by ELISA (A) and TIMP-1, CXCL7, RANTES, L-selectin and sICAM-1 levels measured by protein array at P21 in the rat lung tissue (B,C). NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. Data; mean±SEM. *P<0.05 compared to NC, #P<0.05 compared to HC,†P<0.05 compared to HT3, ‡P<0.05 compared to HT10.
Figure 8. Inflammation with fibrosis in the…
Figure 8. Inflammation with fibrosis in the hyperoxic lung injury after MSCs transplantation.
ED1 positive cells indicative of alveolar macrophage were labeled with FITC (green) and the nuclei were labeled with DAPI (blue) (Scale bar; 25 µm) (at top) and number of observed ED1 positive cells per high power field (below) (A), myeloperoxidase activity (B), and collagen levels (C) of the rat P21 lung tissues. NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. Data; mean±SEM. *P<0.05 compared to NC, #P<0.05 compared to HC,†P<0.05 compared to HT3, ‡P<0.05 compared to HT10.
Figure 9. VEGF and HGF in the…
Figure 9. VEGF and HGF in the hyperoxic lung injury after MSCs transplantation.
Representative RT-PCR blots (at top) and densitometric histograms (below) for HGF (A) and VEGF measured with ELISA (B) in the P21 rat lungs. NC, Normoxia control group; HC, hyperoxia control group; HT3, hyperoxia with stem cell transplantation group at P3; HT10, hyperoxia with stem cell treatment group at P10; HT3+10, hyperoxia with stem cell treatment group at P3 and P10. Data; mean±SEM. *P<0.05 compared to NC, #P<0.05 compared to HC,†P<0.05 compared to HT3, ‡P<0.05 compared to HT10.

References

    1. Saigal S, Doyle LW (2008) An overview of mortality and sequelae of preterm birth from infancy to adulthood. Lancet 371: 261–269.
    1. Avery ME, Tooley WH, Keller JB, Hurd SS, Bryan MH, et al. (1987) Is chronic lung disease in low birth weight infants preventable? A survey of eight centers. Pediatrics 79: 26–30.
    1. Bregman J, Farrell EE (1992) Neurodevelopmental outcome in infants with bronchopulmonary dysplasia. Clin Perinatol 19: 673–694.
    1. Fahy JV (2009) Eosinophilic and neutrophilic inflammation in asthma: insights from clinical studies. Proc Am Thorac Soc 6: 256–259.
    1. Choo-Wing R, Nedrelow JH, Homer RJ, Elias JA, Bhandari V (2007) Developmental differences in the responses of IL-6 and IL-13 transgenic mice exposed to hyperoxia. Am J Physiol Lung Cell Mol Physiol 293: L142–150.
    1. Warner BB, Stuart LA, Papes RA, Wispe JR (1998) Functional and pathological effects of prolonged hyperoxia in neonatal mice. Am J Physiol 275: L110–117.
    1. Chang YS, Oh W, Choi SJ, Sung DK, Kim SY, et al. (2009) Human umbilical cord blood-derived mesenchymal stem cells attenuate hyperoxia-induced lung injury in neonatal rats. Cell Transplant 18: 869–886.
    1. Chang YS, Choi SJ, Sung DK, Kim SY, Oh W, et al. (2011) Intratracheal transplantation of human umbilical cord blood derived mesenchymal stem cells dose-dependently attenuates hyperoxia-induced lung injury in neonatal rats. Cell Transplant
    1. Jang YK, Jung DH, Jung MH, Kim DH, Yoo KH, et al. (2006) Mesenchymal stem cells feeder layer from human umbilical cord blood for ex vivo expanded growth and proliferation of hematopoietic progenitor cells. Ann Hematol 85: 212–225.
    1. Yang SE, Ha CW, Jung M, Jin HJ, Lee M, et al. (2004) Mesenchymal stem/progenitor cells developed in cultures from UC blood. Cytotherapy 6: 476–486.
    1. Boiani M, Scholer HR (2005) Regulatory networks in embryo-derived pluripotent stem cells. Nat Rev Mol Cell Biol 6: 872–884.
    1. Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RC (2007) SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood 109: 1743–1751.
    1. Lee JK, Lee MK, Jin HJ, Kim DS, Yang YS, et al. (2007) Efficient intracytoplasmic labeling of human umbilical cord blood mesenchymal stromal cells with ferumoxides. Cell Transplant 16: 849–857.
    1. Thurlbeck WM (1982) Postnatal human lung growth. Thorax 37: 564–571.
    1. Cho SJ, George CL, Snyder JM, Acarregui MJ (2005) Retinoic acid and erythropoietin maintain alveolar development in mice treated with an angiogenesis inhibitor. Am J Respir Cell Mol Biol 33: 622–628.
    1. Snyder JM, Jenkins-Moore M, Jackson SK, Goss KL, Dai HH, et al. (2005) Alveolarization in retinoic acid receptor-beta-deficient mice. Pediatr Res 57: 384–391.
    1. Babior BM (1999) NADPH oxidase: an update. Blood 93: 1464–1476.
    1. Cross AR, Segal AW (2004) The NADPH oxidase of professional phagocytes–prototype of the NOX electron transport chain systems. Biochim Biophys Acta 1657: 1–22.
    1. Gray KD, Simovic MO, Chapman WC, Blackwell TS, Christman JW, et al. (2003) Endotoxin potentiates lung injury in cerulein-induced pancreatitis. Am J Surg 186: 526–530.
    1. Robinson JM, Badwey JA (1995) The NADPH oxidase complex of phagocytic leukocytes: a biochemical and cytochemical view. Histochem Cell Biol 103: 163–180.
    1. Hohler B, Holzapfel B, Kummer W (2000) NADPH oxidase subunits and superoxide production in porcine pulmonary artery endothelial cells. Histochem Cell Biol 114: 29–37.
    1. Li JM, Shah AM (2002) Intracellular localization and preassembly of the NADPH oxidase complex in cultured endothelial cells. J Biol Chem 277: 19952–19960.
    1. Coalson JJ (2003) Pathology of new bronchopulmonary dysplasia. Semin Neonatol 8: 73–81.
    1. Jobe AJ (1999) The new BPD: an arrest of lung development. Pediatr Res 46: 641–643.
    1. Kunig AM, Balasubramaniam V, Markham NE, Morgan D, Montgomery G, et al. (2005) Recombinant human VEGF treatment enhances alveolarization after hyperoxic lung injury in neonatal rats. Am J Physiol Lung Cell Mol Physiol 289: L529–535.
    1. McGrath-Morrow SA, Stahl J (2001) Apoptosis in neonatal murine lung exposed to hyperoxia. Am J Respir Cell Mol Biol 25: 150–155.
    1. Reinecke H, Murry CE (2002) Taking the death toll after cardiomyocyte grafting: a reminder of the importance of quantitative biology. J Mol Cell Cardiol 34: 251–253.
    1. Togel F, Hu Z, Weiss K, Isaac J, Lange C, et al. (2005) Administered mesenchymal stem cells protect against ischemic acute renal failure through differentiation-independent mechanisms. Am J Physiol Renal Physiol 289: F31–42.
    1. Gnecchi M, He H, Noiseux N, Liang OD, Zhang L, et al. (2006) Evidence supporting paracrine hypothesis for Akt-modified mesenchymal stem cell-mediated cardiac protection and functional improvement. FASEB J 20: 661–669.
    1. Aslam M, Baveja R, Liang OD, Fernandez-Gonzalez A, Lee C, et al. (2009) Bone marrow stromal cells attenuate lung injury in a murine model of neonatal chronic lung disease. Am J Respir Crit Care Med 180: 1122–1130.
    1. Cho HJ, Lee N, Lee JY, Choi YJ, Ii M, et al. (2007) Role of host tissues for sustained humoral effects after endothelial progenitor cell transplantation into the ischemic heart. J Exp Med 204: 3257–3269.
    1. Saugstad OD (2003) Bronchopulmonary dysplasia-oxidative stress and antioxidants. Semin Neonatol 8: 39–49.
    1. Bastian NR, Hibbs JB Jr (1994) Assembly and regulation of NADPH oxidase and nitric oxide synthase. Curr Opin Immunol 6: 131–139.
    1. Moodley Y, Atienza D, Manuelpillai U, Samuel CS, Tchongue J, et al. (2009) Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol 175: 303–313.
    1. Deuse T, Peter C, Fedak PW, Doyle T, Reichenspurner H, et al. (2009) Hepatocyte growth factor or vascular endothelial growth factor gene transfer maximizes mesenchymal stem cell-based myocardial salvage after acute myocardial infarction. Circulation 120: S247–254.
    1. Kokaia Z, Martino G, Schwartz M, Lindvall O (2012) Cross-talk between neural stem cells and immune cells: the key to better brain repair? Nat Neurosci 15: 1078–1087.

Source: PubMed

3
Abonnieren