Mesenchymal stem cell-mediated reversal of bronchopulmonary dysplasia and associated pulmonary hypertension

Georg Hansmann, Angeles Fernandez-Gonzalez, Muhammad Aslam, Sally H Vitali, Thomas Martin, S Alex Mitsialis, Stella Kourembanas, Georg Hansmann, Angeles Fernandez-Gonzalez, Muhammad Aslam, Sally H Vitali, Thomas Martin, S Alex Mitsialis, Stella Kourembanas

Abstract

Clinical trials have failed to demonstrate an effective preventative or therapeutic strategy for bronchopulmonary dysplasia (BPD), a multifactorial chronic lung disease in preterm infants frequently complicated by pulmonary hypertension (PH). Mesenchymal stem cells (MSCs) and their secreted components have been shown to prevent BPD and pulmonary fibrosis in rodent models. We hypothesized that treatment with conditioned media (CM) from cultured mouse bone marrow-derived MSCs could reverse hyperoxia-induced BPD and PH. Newborn mice were exposed to hyperoxia (FiO(2)=0.75) for two weeks, were then treated with one intravenous dose of CM from either MSCs or primary mouse lung fibroblasts (MLFs), and placed in room air for two to four weeks. Histological analysis of lungs harvested at four weeks of age was performed to determine the degree of alveolar injury, blood vessel number, and vascular remodeling. At age six weeks, pulmonary artery pressure (PA acceleration time) and right ventricular hypertrophy (RVH; RV wall thickness) were assessed by echocardiography, and pulmonary function tests were conducted. When compared to MLF-CM, a single dose of MSC-CM-treatment (1) reversed the hyperoxia-induced parenchymal fibrosis and peripheral PA devascularization (pruning), (2) partially reversed alveolar injury, (3) normalized lung function (airway resistance, dynamic lung compliance), (4) fully reversed the moderate PH and RVH, and (5) attenuated peripheral PA muscularization associated with hyperoxia-induced BPD. Reversal of key features of hyperoxia-induced BPD and its long-term adverse effects on lung function can be achieved by a single intravenous dose of MSC-CM, thereby pointing toward a new therapeutic intervention for chronic lung diseases.

Keywords: airway hyperresponsiveness; chronic lung disease of infancy; hyperoxia; lung injury; lung vascular pruning.

Conflict of interest statement

Conflict of Interest: None declared.

Figures

Figure 1
Figure 1
MSC-CM-treatment partially reverses neonatal hyperoxia-induced alveolar injury, septal thickening, collagen accumulation, myofibroblast infiltration, and inflammation. Upper panel: Experimental Design. Newborn mice were either left in room air, or exposed to hyperoxia (FiO2=0.75) for 2 weeks (P1-14), and then intravenously injected once with CM from either bone marrow-derived MSCs or primary MLFs, and placed in room air for 2 additional weeks, followed by harvest, inflation and fixation of lungs. Lung sections were stained for hemotoxylin & eosin (A, C, E; 100×), and Mason Trichrome (B, D, F; 400×). Inserts were taken at 200× (H&E) and 400× (Mason Trichrome) magnification to illustrate interstitial inflammation (C, arrows), inflammation and fibrosis (D). Severe destruction of the alveolar architecture with overall widened airspaces and interstitial infiltration of inflammatory cells (macrophages, leukocytes) and myofibroblasts was seen in the hyperoxia-exposed/MLF-CM-treated mice (C, D) when compared to normoxic controls (A, B). Enhanced collagen deposition was seen in myofibroblasts, alveolar septa, and perivascular spaces in hyperoxia-exposed/MLF-CM-treated mouse lungs (D, arrows and inset). These changes were absent or greatly abrogated in MSC-CM-treated mice that had honeycomb-like alveoli (E, F) similar to normoxic controls, with residual emphysema. Quantification of mean linear intercept (Lm) as a surrogate of average air space diameter (G) and collagen content (H) are shown in the lower panels. Mean±SEM, n=4-7 per group, ***P<0.001, **P<0.01, *P<0.05. Scale bar = 100 μm (A, C, E) and 50 μm (B, D, F).
Figure 2
Figure 2
MSC-CM improve lung function after hyperoxia-induced lung injury. Pulmonary function testing was performed in six-week-old mice four weeks after the end of hyperoxia, and in age-matched normoxic control mice (see experimental design shown in Figure 1, and Methods section). Airway resistance (A) was measured at baseline and under escalating doses of intratracheal methacholine in order to quantify bronchial hyperreactivity. At baseline, airway resistance was not different between the three groups. MSC-CM-treatment fully reversed the abnormal increase in airway resistance seen in the MLF-CM group after administration of low to high intratracheal methacholine doses (5, 16, 50 mg/ml), to levels not different from normoxic controls (A). Dynamic lung compliance (Cdyn) was remarkably impaired in the hyperoxia-exposed/MLF-CM-treated mice at a methacholine dose of 50 mg/ml but normal in the MSC-CM-treated mice when compared with normoxic controls, indicating normalized compliance under metacholine stress (B). The corresponding histological findings are shown in Figure 1. Mean±SEM, n=3–4 per group, ** P<0.01, * P<0.05.
Figure 3
Figure 3
MSC-CM-treatment reverses pulmonary hypertension and RV hypertrophy in hyperoxia-induced lung injury. Pulmonary artery acceleration time (PAAT; syn. PAT), as a surrogate of mean PA pressure, was echocardiographically measured by PW-Doppler (A, C, E) and found to be shortened in hyperoxia-exposed/MLF-CM-treated animals (C), but normal in hyperoxia-exposed/MSC-CM-treated mice (blue line, E), when compared to normoxic controls (A, graphically summarized in G). Similar results were obtained for the PAAT/PAET ratio, where PAET is the pulmonary artery ejection time (syn. PET; data not shown). The end-diastolic diameter of the RV free wall (RVWT, RV wall thickness) was measured by M-mode echocardiography (B, D, F) and found to be increased in the hyperoxia-exposed/MLF-CM-treated mice but normalized in the MSC-CM-treated animals (H). Thus, a single dose of MSC-CM reversed the moderate pulmonary hypertension (G) and RV hypertrophy (H) that was associated with hyperoxia-induced BPD. See Figure 1 for experimental design. Mean±SEM, n=4 per group, * P<0.05.
Figure 4
Figure 4
MSC-CM attenuate hyperoxia-induced peripheral pulmonary arteriole remodeling. Lung sections were stained for the smooth muscle marker, α-SMA, and peripheral PA muscularization (P<0.05. Scale bar=25 μm.
Figure 5
Figure 5
MSC-CM rescue hyperoxia-induced loss of peripheral pulmonary blood vessels. Lung sections were stained for the endothelial cell marker vWF. vWF-positive vessels between 25 and 200 μm outer diameter were counted at 200× magnification in 10–15 random views as described under Methods. Compared to normoxic controls (A), hyperoxia exposure led to significant loss of small vessels P=0.0534). Quantification of pulmonary blood vessels of less than 50 and 50–100 μm diameter in vWF-stained lung sections (D) was performed as described under Methods. There was no significant difference in numbers of larger vessels (100-200 μm) between the groups. See Figure 1 for experimental design. Mean±SEM, n=4–7 per group, ** P<0.01, * P<0.05. Scale bar = 50 μm.
Figure 6
Figure 6
MSC-CM reverse pulmonary artery pruning in hyperoxia-induced lung injury. Pulmonary artery barium injections and subsequent computed tomography angiograms with 3D reconstruction of the PA vascularization of left lungs were performed ex vivo, as described in the Methods section. When compared to normoxic controls (A), severe rarefication (pruning) of peripheral PAs (arrows) was evident on CT-angiograms of hyperoxia-exposed/MLF-CM-treated mice at 4 weeks recovery in room air (B). Such hyperoxia-induced PA pruning was completely absent in lungs from mice at 4 weeks recovery in room air injected with a single intravenous dose of MSC-CM at the end of hyperoxia exposure (C). For experimental design, see Figure 1.

References

    1. Jobe AH, Bancalari E. Bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2001;163:1723–9.
    1. Stenmark KR, Abman SH. Lung vascular development: implications for the pathogenesis of bronchopulmonary dysplasia. Annu Rev Physiol. 2005;67:623–61.
    1. Thebaud B, Abman SH. Bronchopulmonary dysplasia: Where have all the vessels gone? Roles of angiogenic growth factors in chronic lung disease. Am J Respir Crit Care Med. 2007;175:978–85.
    1. Baraldi E, Filippone M. Chronic lung disease after premature birth. N Engl J Med. 2007;357:1946–55.
    1. Kim DH, Kim HS, Choi CW, Kim EK, Kim BI, Choi JH. Risk factors for pulmonary artery hypertension in preterm infants with moderate or severe bronchopulmonary dysplasia. Neonatology. 2012;101:40–6.
    1. Khemani E, McElhinney DB, Rhein L, Andrade O, Lacro RV, Thomas KC, et al. Pulmonary artery hypertension in formerly premature infants with bronchopulmonary dysplasia: Clinical features and outcomes in the surfactant era. Pediatrics. 2007;120:1260–9.
    1. Cerro MJ, Abman S, Diaz G, Freudenthal AH, Freudenthal F, Harikrishnan S, et al. A consensus approach to the classification of pediatric pulmonary hypertensive vascular disease: Report from the PVRI Pediatric Taskforce, Panama 2011. Pulm Circ. 2011;1:286–98.
    1. Warner BB, Stuart LA, Papes RA, Wispe JR. Functional and pathological effects of prolonged hyperoxia in neonatal mice. Am J Physiol. 1998;275:L110–L7.
    1. Ortiz LA, Gambelli F, McBride C, Gaupp D, Baddoo M, Kaminski N, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proc Natl Acad Sci U S A. 2003;100:8407–11.
    1. Weiss DJ, Bertoncello I, Borok Z, Kim C, Panoskaltsis-Mortari A, Reynolds S, et al. Stem cells and cell therapies in lung biology and lung diseases. Proc Am Thorac Soc. 2011;8:223–72.
    1. Aslam M, Baveja R, Liang OD, Fernandez-Gonzalez A, Lee C, Mitsialis SA, et al. Bone marrow stromal cells attenuate lung injury in a murine model of neonatal chronic lung disease. Am J Respir Crit Care Med. 2009;180:1122–30.
    1. van Haaften T, Byrne R, Bonnet S, Rochefort GY, Akabutu J, Bouchentouf M, et al. Airway delivery of mesenchymal stem cells prevents arrested alveolar growth in neonatal lung injury in rats. Am J Respir Crit Care Med. 2009;180:1131–42.
    1. Gupta N, Su X, Popov B, Lee JW, Serikov V, Matthay MA. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol. 2007;179:1855–63.
    1. Mei SH, McCarter SD, Deng Y, Parker CH, Liles WC, Stewart DJ. Prevention of LPS-induced acute lung injury in mice by mesenchymal stem cells overexpressing angiopoietin 1. PLoS Med. 2007;4:e269.
    1. Saugstad OD. Resuscitation of newborn infants: From oxygen to room air. Lancet. 2010;376:1970–1.
    1. Ozdemir R, Erdeve O, Dizdar EA, Oguz SS, Uras N, Saygan S, et al. Clarithromycin in preventing bronchopulmonary dysplasia in Ureaplasma urealyticum-positive preterm infants. Pediatrics. 2011;128:e1496–501.
    1. Gopel W, Kribs A, Ziegler A, Laux R, Hoehn T, Wieg C, et al. Avoidance of mechanical ventilation by surfactant treatment of spontaneously breathing preterm infants (AMV): An open-label, randomised, controlled trial. Lancet. 2011;378:1627–34.
    1. Gien J, Kinsella JP. Pathogenesis and treatment of bronchopulmonary dysplasia. Curr Opin Pediatr. 2011;23:305–13.
    1. Hansen AR, Barnes CM, Folkman J, McElrath TF. Maternal preeclampsia predicts the development of bronchopulmonary dysplasia. J Pediatr. 2010;156:532–6.
    1. Tang JR, Karumanchi SA, Seedorf G, Markham N, Abman SH. Excess soluble vascular endothelial growth factor receptor-1 in amniotic fluid impairs lung growth in rats: Linking preeclampsia with bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2012;302:L36–46.
    1. Watterberg KL, Demers LM, Scott SM, Murphy S. Chorioamnionitis and early lung inflammation in infants in whom bronchopulmonary dysplasia develops. Pediatrics. 1996;97:210–5.
    1. Paananen R, Husa AK, Vuolteenaho R, Herva R, Kaukola T, Hallman M. Blood cytokines during the perinatal period in very preterm infants: Relationship of inflammatory response and bronchopulmonary dysplasia. J Pediatr. 2009;154:39–43. e3.
    1. Hansmann G. Neonatal resuscitation on air: It is time to turn down the oxygen tanks [corrected] Lancet. 2004;364:1293–4.
    1. Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell. 2008;3:301–13.
    1. Mangi AA, Noiseux N, Kong D, He H, Rezvani M, Ingwall JS, et al. Mesenchymal stem cells modified with Akt prevent remodeling and restore performance of infarcted hearts. Nat Med. 2003;9:1195–201.
    1. Peister A, Mellad JA, Larson BL, Hall BM, Gibson LF, Prockop DJ. Adult stem cells from bone marrow (MSCs) isolated from different strains of inbred mice vary in surface epitopes, rates of proliferation, and differentiation potential. Blood. 2004;103:1662–8.
    1. Dobson KR, Reading L, Haberey M, Marine X, Scutt A. Centrifugal isolation of bone marrow from bone: An improved method for the recovery and quantitation of bone marrow osteoprogenitor cells from rat tibiae and femurae. Calcif Tissue Int. 1999;65:411–3.
    1. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells.The International Society for Cellular Therapy position statement. Cytotherapy. 2006;8:315–7.
    1. McGowan SE. Influences of endogenous and exogenous TGF-beta on elastin in rat lung fibroblasts and aortic smooth muscle cells. Am J Physiol. 1992;263:L257–63.
    1. Thibault HB, Kurtz B, Raher MJ, Shaik RS, Waxman A, Derumeaux G, et al. Noninvasive assessment of murine pulmonary arterial pressure: Validation and application to models of pulmonary hypertension. Circ Cardiovasc Imaging. 2010;3:157–63.
    1. Urboniene D, Haber I, Fang YH, Thenappan T, Archer SL. Validation of high-resolution echocardiography and magnetic resonance imaging vs.high-fidelity catheterization in experimental pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2010;299:L401–12.
    1. Hansmann G, Wagner RA, Schellong S, Perez VA, Urashima T, Wang L, et al. Pulmonary arterial hypertension is linked to insulin resistance and reversed by peroxisome proliferator-activated receptor-gamma activation. Circulation. 2007;115:1275–84.
    1. Siniscalco D, Sullo N, Maione S, Rossi F, D’Agostino B. Stem cell therapy: The great promise in lung disease. Ther Adv Respir Dis. 2008;2:173–7.
    1. McQualter JL, Bertoncello I. Concise Review: Deconstructing the lung to reveal its regenerative potential. Stem Cells. 2012;30:811–6.
    1. Williams AR, Hare JM. Mesenchymal stem cells: Biology, pathophysiology, translational findings, and therapeutic implications for cardiac disease. Circ Res. 2011;109:923–40.
    1. Lee JW, Fang X, Krasnodembskaya A, Howard JP, Matthay MA. Concise review: Mesenchymal stem cells for acute lung injury: Role of paracrine soluble factors. Stem Cells. 2011;29:913–9.
    1. Chang YS, Oh W, Choi SJ, Sung DK, Kim SY, Choi EY, et al. Human umbilical cord blood-derived mesenchymal stem cells attenuate hyperoxia-induced lung injury in neonatal rats. Cell Transplant. 2009;18:869–86.
    1. Tropea KA, Leder E, Aslam M, Lau AN, Raiser DM, Lee JH, et al. Bronchioalveolar stem cells increase after mesenchymal stromal cell treatment in a mouse model of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2012;302:L829–L837.
    1. Augello A, De Bari C. The regulation of differentiation in mesenchymal stem cells. Hum Gene Ther. 2010;21:1226–38.
    1. Popova AP, Bozyk PD, Goldsmith AM, Linn MJ, Lei J, Bentley JK, et al. Autocrine production of TGF-beta1 promotes myofibroblastic differentiation of neonatal lung mesenchymal stem cells. Am J Physiol Lung Cell Mol Physiol. 2010;298:L735–43.
    1. Dasgupta C, Sakurai R, Wang Y, Guo P, Ambalavanan N, Torday JS, et al. Hyperoxia-induced neonatal rat lung injury involves activation of TGF-{beta} and Wnt signaling and is protected by rosiglitazone. Am J Physiol Lung Cell Mol Physiol. 2009;296:L1031–41.
    1. Kumarasamy A, Schmitt I, Nave AH, Reiss I, van der Horst I, Dony E, et al. Lysyl oxidase activity is dysregulated during impaired alveolarization of mouse and human lungs. Am J Respir Crit Care Med. 2009;180:1239–52.
    1. Nakanishi H, Sugiura T, Streisand JB, Lonning SM, Roberts JD., Jr TGF-beta-neutralizing antibodies improve pulmonary alveologenesis and vasculogenesis in the injured newborn lung. Am J Physiol Lung Cell Mol Physiol. 2007;293:L151–61.
    1. Konigshoff M, Kramer M, Balsara N, Wilhelm J, Amarie OV, Jahn A, et al. WNT1-inducible signaling protein-1 mediates pulmonary fibrosis in mice and is upregulated in humans with idiopathic pulmonary fibrosis. J Clin Invest. 2009;119:772–87.
    1. Henderson WR, Jr, Chi EY, Ye X, Nguyen C, Tien YT, Zhou B, et al. Inhibition of Wnt/beta-catenin/CREB binding protein (CBP) signaling reverses pulmonary fibrosis. Proc Natl Acad Sci U S A. 2010;107:14309–14.
    1. Wynn TA. Integrating mechanisms of pulmonary fibrosis. J Exp Med. 2011;208:1339–50.
    1. da Silva Meirelles L, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci. 2006;119:2204–13.
    1. Zannettino AC, Paton S, Arthur A, Khor F, Itescu S, Gimble JM, et al. Multipotential human adipose-derived stromal stem cells exhibit a perivascular phenotype in vitro and in vivo. J Cell Physiol. 2008;214:413–21.
    1. Popova AP, Bozyk PD, Bentley JK, Linn MJ, Goldsmith AM, Schumacher RE, et al. Isolation of tracheal aspirate mesenchymal stromal cells predicts bronchopulmonary dysplasia. Pediatrics. 2010;126:e1127–33.
    1. Balasubramaniam V, Mervis CF, Maxey AM, Markham NE, Abman SH. Hyperoxia reduces bone marrow, circulating, and lung endothelial progenitor cells in the developing lung: implications for the pathogenesis of bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol. 2007;292:L1073–84.
    1. Baker CD, Ryan SL, Ingram DA, Seedorf GJ, Abman SH, Balasubramaniam V. Endothelial colony-forming cells from preterm infants are increased and more susceptible to hyperoxia. Am J Respir Crit Care Med. 2009;180:454–61.
    1. Borghesi A, Massa M, Campanelli R, Bollani L, Tzialla C, Figar TA, et al. Circulating endothelial progenitor cells in preterm infants with bronchopulmonary dysplasia. Am J Respir Crit Care Med. 2009;180:540–6.
    1. Minamino K, Adachi Y, Okigaki M, Ito H, Togawa Y, Fujita K, et al. Macrophage colony-stimulating factor (M-CSF), as well as granulocyte colony-stimulating factor (G-CSF), accelerates neovascularization. Stem Cells. 2005;23:347–54.
    1. Zhang Y, Adachi Y, Iwasaki M, Minamino K, Suzuki Y, Nakano K, et al. G-CSF and/or M-CSF accelerate differentiation of bone marrow cells into endothelial progenitor cells in vitro. Oncol Rep. 2006;15:1523–7.
    1. Nakano K, Adachi Y, Minamino K, Iwasaki M, Shigematsu A, Kiriyama N, et al. Mechanisms underlying acceleration of blood flow recovery in ischemic limbs by macrophage colony-stimulating factor. Stem Cells. 2006;24:1274–9.
    1. Moore BB, Kolodsick JE, Thannickal VJ, Cooke K, Moore TA, Hogaboam C, et al. CCR2-mediated recruitment of fibrocytes to the alveolar space after fibrotic injury. Am J Pathol. 2005;166:675–84.
    1. Frid MG, Brunetti JA, Burke DL, Carpenter TC, Davie NJ, Reeves JT, et al. Hypoxia-induced pulmonary vascular remodeling requires recruitment of circulating mesenchymal precursors of a monocyte/macrophage lineage. Am J Pathol. 2006;168:659–69.

Source: PubMed

3
Abonnieren