Hematopoietic and mesenchymal stem cells for the treatment of chronic respiratory diseases: role of plasticity and heterogeneity

Massimo Conese, Donatella Piro, Annalucia Carbone, Stefano Castellani, Sante Di Gioia, Massimo Conese, Donatella Piro, Annalucia Carbone, Stefano Castellani, Sante Di Gioia

Abstract

Chronic lung diseases, such as cystic fibrosis (CF), asthma, and chronic obstructive pulmonary disease (COPD) are incurable and represent a very high social burden. Stem cell-based treatment may represent a hope for the cure of these diseases. In this paper, we revise the overall knowledge about the plasticity and engraftment of exogenous marrow-derived stem cells into the lung, as well as their usefulness in lung repair and therapy of chronic lung diseases. The lung is easily accessible and the pathophysiology of these diseases is characterized by injury, inflammation, and eventually by remodeling of the airways. Bone marrow-derived stem cells, including hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stromal (stem) cells (MSCs), encompass a wide array of cell subsets with different capacities of engraftment and injured tissue regenerating potential. Proof-of-principle that marrow cells administered locally may engraft and give rise to specialized epithelial cells has been given, but the efficiency of this conversion is too limited to give a therapeutic effect. Besides the identification of plasticity mechanisms, the characterization/isolation of the stem cell subpopulations represents a major challenge to improving the efficacy of transplantation protocols used in regenerative medicine for lung diseases.

Figures

Figure 1
Figure 1
Still debated issues about homing and engraftment of marrow cells in the lung.

References

    1. Murray CJL, Lopez AD. Alternative projections of mortality and disability by cause 1990–2020: global Burden of Disease Study. The Lancet. 1997;349(9064):1498–1504.
    1. Barnes PJ. Corticosteroid resistance in patients with asthma and chronic obstructive pulmonary disease. Journal of Allergy and Clinical Immunology. 2013;131(3):636–645.
    1. Becq F. Cystic fibrosis transmembrane conductance regulator modulators for personalized drug treatment of cystic fibrosis: progress to date. Drugs. 2010;70(3):241–259.
    1. Grove JE, Bruscia E, Krause DS. Plasticity of bone marrow-derived stem cells. Stem Cells. 2004;22(4):487–500.
    1. Denburg JA, van Eeden SF. Bone marrow progenitors in inflammation and repair: new vistas in respiratory biology and pathophysiology. European Respiratory Journal. 2006;27(3):441–445.
    1. Weiss DJ, Berberich MA, Borok Z, et al. Adult stem cells, lung biology, and lung disease. Proceedings of the American Thoracic Society. 2006;3(3):193–207.
    1. Gomperts BN, Strieter RM. Stem cells and chronic lung disease. Annual Review of Medicine. 2007;58:285–298.
    1. Weiss DJ, Kolls JK, Ortiz LA, Panoskaltsis-Mortari A, Prockop DJ. Stem cells and cell therapies in lung biology and lung diseases. Proceedings of the American Thoracic Society. 2008;5(5):637–667.
    1. Sueblinvong V, Weiss DJ. Stem cells and cell therapy approaches in lung biology and diseases. Translational Research. 2010;156(3):188–205.
    1. Pesci A, Balbi B, Majori M, et al. Inflammatory cells and mediators in bronchial lavage of patients with chronic obstructive pulmonary disease. European Respiratory Journal. 1998;12(2):380–386.
    1. Barnes PJ. Chronic obstructive pulmonary disease. The New England Journal of Medicine. 2000;343(4):269–280.
    1. Fischer BM, Pavlisko E, Voynow JA. Pathogenic triad in COPD: oxidative stress, protease-antiprotease imbalance, and inflammation. International Journal of Chronic Obstructive Pulmonary Disease. 2011;6:413–421.
    1. Sahiner UM, Birben E, Erzurum S, Sackesen C, Kalayci O. Oxidative stress in asthma. World Allergy Organization Journal. 2011;4(10):151–158.
    1. Barnes PJ. Pathophysiology of allergic inflammation. Immunological Reviews. 2011;242(1):31–50.
    1. Aoshiba K, Nagai A. Differences in airway remodeling between asthma and chronic obstructive pulmonary disease. Clinical Reviews in Allergy and Immunology. 2004;27(1):35–43.
    1. Akabas MH. Cystic fibrosis transmembrane conductance regulator. Structure and function of an epithelial chloride channel. Journal of Biological Chemistry. 2000;275(6):3729–3732.
    1. Boucher RC. Cystic fibrosis: a disease of vulnerability to airway surface dehydration. Trends in Molecular Medicine. 2007;13(6):231–240.
    1. Rowe SM, Miller S, Sorscher EJ. Cystic fibrosis. The New England Journal of Medicine. 2005;352(19):1992–2001.
    1. Hartl D, Latzin P, Hordijk P, et al. Cleavage of CXCR1 on neutrophils disables bacterial killing in cystic fibrosis lung disease. Nature Medicine. 2007;13(12):1423–1430.
    1. Conese M. Cystic fibrosis and the innate immune system: therapeutic implications. Endocrine, Metabolic and Immune Disorders. 2011;11(1):8–22.
    1. Di A, Brown ME, Deriy LV, et al. CFTR regulates phagosome acidification in macrophages and alters bactericidal activity. Nature Cell Biology. 2006;8(9):933–944.
    1. Zhang Y, Li X, Grassmé H, Döring G, Gulbins E. Alterations in ceramide concentration and pH determine the release of reactive oxygen species by Cftr-deficient macrophages on infection. Journal of Immunology. 2010;184(9):5104–5111.
    1. Hilliard TN, Regamey N, Shute JK, et al. Airway remodelling in children with cystic fibrosis. Thorax. 2007;62(12):1074–1080.
    1. Ulrich M, Worlitzsch D, Viglio S, et al. Alveolar inflammation in cystic fibrosis. Journal of Cystic Fibrosis. 2010;9(3):217–227.
    1. Gaggar A, Hector A, Bratcher PE, Mall MA, Griese M, Hartl D. Series “matrix metalloproteinases in lung health and disease”: the role of matrix metalloproteinases in cystic fibrosis lung disease. European Respiratory Journal. 2011;38(3):721–727.
    1. Krause DS, Theise ND, Collector MI, et al. Multi-organ, multi-lineage engraftment by a single bone marrow-derived stem cell. Cell. 2001;105(3):369–377.
    1. Wong AP, Dutly AE, Sacher A, et al. Targeted cell replacement with bone marrow cells for airway epithelial regeneration. American Journal of Physiology—Lung Cellular and Molecular Physiology. 2007;293(3):L740–L752.
    1. Tropea KA, Leder E, Aslam M, et al. Bronchioalveolar stem cells increase after mesenchymal stromal cell treatment in a mouse model of bronchopulmonary dysplasia. American Journal of Physiology—Lung Cellular and Molecular Physiology. 2012;302(9):L829–L837.
    1. Eisenhauer P, Earle B, Loi R, et al. Endogenous distal airway progenitor cells, lung mechanics, and disproportionate lobar growth following long-term postpneumonectomy in mice. Stem Cells. 2013;31(7):1330–1339.
    1. Abe S, Boyer C, Liu X, et al. Cells derived from the circulation contribute to the repair of lung injury. American Journal of Respiratory and Critical Care Medicine. 2004;170(11):1158–1163.
    1. Theise ND, Henegariu O, Grove J, et al. Radiation pneumonitis in mice: a severe injury model for pneumocyte engraftment from bone marrow. Experimental Hematology. 2002;30(11):1333–1338.
    1. Grove JE, Lutzko C, Priller J, et al. Marrow-derived cells as vehicles for delivery of gene therapy to pulmonary epithelium. American Journal of Respiratory Cell and Molecular Biology. 2002;27(6):645–651.
    1. Serikov VB, Popov B, Mikhailov VM, Gupta N, Matthay MA. Evidence of temporary airway epithelial repopulation and rare clonal formation by BM-derived cells following naphthalene injury in mice. Anatomical Record. 2007;290(9):1033–1045.
    1. Suratt BT, Cool CD, Serls AE, et al. Human pulmonary chimerism after hematopoietic stem cell transplantation. American Journal of Respiratory and Critical Care Medicine. 2003;168(3):318–322.
    1. Mattsson J, Jansson M, Wernerson A, Hassan M. Lung epithelial cells and type II pneumocytes of donor origin after allogeneic hematopoietic stem cell tansplantation. Transplantation. 2004;78(1):154–157.
    1. Kleeberger W, Versmold A, Rothämel T, et al. Increased chimerism of bronchial and alveolar epithelium in human lung allografts undergoing chronic injury. American Journal of Pathology. 2003;162(5):1487–1494.
    1. Weiss DJ, Bertoncello I, Borok Z, et al. Stem cells and cell therapies in lung biology and lung diseases. Proceedings of the American Thoracic Society. 2011;8(3):223–272.
    1. Aliotta JM, Keaney P, Passero M, et al. Bone marrow production of lung cells: the impact of G-CSF, cardiotoxin, graded doses of irradiation, and subpopulation phenotype. Experimental Hematology. 2006;34(2):230–241.
    1. Herzog EL, Van Arnam J, Hu B, Krause DS. Threshold of lung injury required for the appearance of marrow-derived lung epithelia. Stem Cells. 2006;24(8):1986–1992.
    1. Conese M, Copreni E, Piro D, Rejman J. Gene and cell therapy for the treatment of cystic fibrosis. Advances in Gene, Molecular and Cell Therapy. 2007;1:99–119.
    1. MacPherson H, Keir PA, Edwards CJ, Webb S, Dorin JR. Following damage, the majority of bone marrow-derived airway cells express an epithelial marker. Respiratory Research. 2006;7, article 145
    1. Rojas M, Xu J, Woods CR, et al. Bone marrow-derived mesenchymal stem cells in repair of the injured lung. American Journal of Respiratory Cell and Molecular Biology. 2005;33(2):145–152.
    1. Kotton DN, Ma BY, Cardoso WV, et al. Bone marrow-derived cells as progenitors of lung alveolar epithelium. Development. 2001;128(24):5181–5188.
    1. Kotton DN, Fabian AJ, Mulligan RC. Failure of bone marrow to reconstitute lung epithelium. American Journal of Respiratory Cell and Molecular Biology. 2005;33(4):328–334.
    1. Wagers AJ, Sherwood RI, Christensen JL, Weissman IL. Little evidence for developmental plasticity of adult hematopoietic stem cells. Science. 2002;297(5590):2256–2259.
    1. Chang JC, Summer R, Sun X, Fitzsimmons K, Fine A. Evidence that bone marrow cells do not contribute to the alveolar epithelium. American Journal of Respiratory Cell and Molecular Biology. 2005;33(4):335–342.
    1. Fritzell JA, Jr., Mao Q, Gundavarapu S, et al. Fate and effects of adult bone marrow cells in lungs of normoxic and hyperoxic newborn mice. American Journal of Respiratory Cell and Molecular Biology. 2009;40(5):575–587.
    1. Quesenberry PJ, Colvin G, Dooner G, Dooner M, Aliotta JM, Johnson K. The stem cell continuum: cell cycle, injury, and phenotype lability. Annals of the New York Academy of Sciences. 2007;1106:20–29.
    1. Krause DS. Bone marrow-derived lung epithelial cells. Proceedings of the American Thoracic Society. 2008;5(6):699–702.
    1. Kassmer SH, Krause DS. Detection of bone marrow-derived lung epithelial cells. Experimental Hematology. 2010;38(7):564–573.
    1. Leblond A-L, Naud P, Forest V, et al. Developing cell therapy techniques for respiratory disease: intratracheal delivery of genetically engineered stem cells in a murine model of airway injury. Human Gene Therapy. 2009;20(11):1329–1343.
    1. Rejman J, Colombo C, Conese M. Engraftment of bone marrow-derived stem cells to the lung in a model of acute respiratory infection by Pseudomonas aeruginosa. Molecular Therapy. 2009;17(7):1257–1265.
    1. Jiang Y, Jahagirdar BN, Reinhardt RL, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002;418(6893):41–49.
    1. Kucia M, Reca R, Jala VR, Dawn B, Ratajczak J, Ratajczak MZ. Bone marrow as a home of heterogenous populations of nonhematopoietic stem cells. Leukemia. 2005;19(7):1118–1127.
    1. Kucia M, Ratajczak J, Ratajczak MZ. Bone marrow as a source of circulating CXCR4+ tissue-committed stem cells. Biology of the Cell. 2005;97(2):133–146.
    1. Gomperts BN, Belperio JA, Rao PN, et al. Circulating progenitor epithelial cells traffic via CXCR4/CXCL12 in response to airway injury. Journal of Immunology. 2006;176(3):1916–1927.
    1. Harris RG, Herzog EL, Bruscia EM, Grove JE, Van Arnam JS, Krause DS. Lack of a fusion requirement for development of bone marrow-derived epithelia. Science. 2004;305(5680):90–93.
    1. Alvarez-Dolado M, Pardal R, Garcia-Verdugo JM, et al. Fusion of bone-marrow-derived cells with Purkinje neurons, cardiomyocytes and hepatocytes. Nature. 2003;425(6961):968–973.
    1. Herzog EL, Van Arnam J, Hu B, et al. Lung-specific nuclear reprogramming is accompanied by heterokaryon formation and Y chromosome loss following bone marrow transplantation and secondary inflammation. FASEB Journal. 2007;21(10):2592–2601.
    1. Aliotta JM, Sanchez-Guijo FM, Dooner GJ, et al. Alteration of marrow cell gene expression, protein production, and engraftment into lung by lung-derived microvesicles: a novel mechanism for phenotype modulation. Stem Cells. 2007;25(9):2245–2256.
    1. Aliotta JM, Pereira M, Johnson KW, et al. Microvesicle entry into marrow cells mediates tissue-specific changes in mRNA by direct delivery of mRNA and induction of transcription. Experimental Hematology. 2010;38(3):233–245.
    1. Lau AN, Goodwin M, Kim CF, Weiss DJ. Stem cells and regenerative medicine in lung biology and diseases. Molecular Therapy. 2012;20(6):1116–1130.
    1. Conese M, Carbone A, Castellani S, Di Gioia S. Paracrine effects and heterogeneity of marrow-derived stem/progenitor cells: relevance for the treatment of respiratory diseases. Cells Tissues Organs. 2013;197(6):445–473.
    1. Yamada M, Kubo H, Kobayashi S, et al. Bone marrow-derived progenitor cells are important for lung repair after lipopolysaccharide-induced lung injury. Journal of Immunology. 2004;172(2):1266–1272.
    1. Wang X-X, Zhang F-R, Shang Y-P, et al. Transplantation of autologous endothelial progenitor cells may be beneficial in patients with idiopathic pulmonary arterial hypertension. A pilot randomized controlled trial. Journal of the American College of Cardiology. 2007;49(14):1566–1571.
    1. Zhu JH, Wang XX, Zhang FR, et al. Safety and efficacy of autologous endothelial progenitor cells transplantation in children with idiopathic pulmonary arterial hypertension: open-label pilot study. Pediatric Transplantation. 2008;12(6):650–655.
    1. Diller G-P, Thum T, Wilkins MR, Wharton J. Endothelial progenitor cells in pulmonary arterial hypertension. Trends in Cardiovascular Medicine. 2010;20(1):22–29.
    1. Phillips RJ, Burdick MD, Hong K, et al. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. Journal of Clinical Investigation. 2004;114(3):438–446.
    1. Epperly MW, Guo H, Gretton JE, Greenberger JS. Bone marrow origin of myofibroblasts in irradiation pulmonary fibrosis. American Journal of Respiratory Cell and Molecular Biology. 2003;29(2):213–224.
    1. Hashimoto N, Jin H, Liu T, Chensue SW, Phan SH. Bone marrow-derived progenitor cells in pulmonary fibrosis. Journal of Clinical Investigation. 2004;113(2):243–252.
    1. Moore BB, Kolodsick JE, Thannickal VJ, et al. CCR2-mediated recruitment of fibrocytes to the alveolar space after fibrotic injury. American Journal of Pathology. 2005;166(3):675–684.
    1. Tanjore H, Xu XC, Polosukhin VV, et al. Contribution of epithelial-derived fibroblasts to bleomycin-induced lung fibrosis. American Journal of Respiratory and Critical Care Medicine. 2009;180(7):657–665.
    1. Strieter RM, Keeley EC, Burdick MD, Mehrad B. The role of circulating mesenchymal progenitor cells, fibrocytes, in promoting pulmonary fibrosis. Transactions of the American Clinical and Climatological Association. 2009;120:49–59.
    1. Keeley EC, Mehrad B, Strieter RM. Fibrocytes: bringing new insights into mechanisms of inflammation and fibrosis. International Journal of Biochemistry and Cell Biology. 2010;42(4):535–542.
    1. Gomperts BN, Strieter RM. Fibrocytes in lung disease. Journal of Leukocyte Biology. 2007;82(3):449–456.
    1. Bellini A, Mattoli S. The role of the fibrocyte, a bone marrow-derived mesenchymal progenitor, in reactive and reparative fibroses. Laboratory Investigation. 2007;87(9):858–870.
    1. Ortiz LA, Gambelli F, McBride C, et al. Mesenchymal stem cell engraftment in lung is enhanced in response to bleomycin exposure and ameliorates its fibrotic effects. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(14):8407–8411.
    1. Gupta N, Su X, Popov B, Jae WL, Serikov V, Matthay MA. Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. Journal of Immunology. 2007;179(3):1855–1863.
    1. Xu J, Woods CR, Mora AL, et al. Prevention of endotoxin-induced systemic response by bone marrow-derived mesenchymal stem cells in mice. American Journal of Physiology—Lung Cellular and Molecular Physiology. 2007;293(1):L131–L141.
    1. Ortiz LA, DuTreil M, Fattman C, et al. Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(26):11002–11007.
    1. Chamberlain G, Fox J, Ashton B, Middleton J. Concise review: mesenchymal stem cells: their phenotype, differentiation capacity, immunological features, and potential for homing. Stem Cells. 2007;25(11):2739–2749.
    1. Rasmusson I. Immune modulation by mesenchymal stem cells. Experimental Cell Research. 2006;312(12):2169–2179.
    1. Duffy MM, Ritter T, Ceredig R, Griffin MD. Mesenchymal stem cell effects on T-cell effector pathways. Stem Cell Research and Therapy. 2011;2(4, article 34)
    1. Bifari F, Lisi V, Mimiola E, Pasini A, Krampera M. Immune modulation by mesenchymal stem cells. Transfusion Medicine and Hemotherapy. 2008;35(3):194–204.
    1. Le Blanc K, Frassoni F, Ball L, et al. Mesenchymal stem cells for treatment of steroid-resistant, severe, acute graft-versus-host disease: a phase II study. The Lancet. 2008;371(9624):1579–1586.
    1. Duijvestein M, Vos ACW, Roelofs H, et al. Autologous bone marrow-derived mesenchymal stromal cell treatment for refractory luminal Crohn’s disease: results of a phase I study. Gut. 2010;59(12):1662–1669.
    1. Salem HK, Thiemermann C. Mesenchymal stromal cells: current understanding and clinical status. Stem Cells. 2010;28(3):585–596.
    1. Abreu SC, Antunes MA, Pelosi P, Morales MM, Rocco PRM. Mechanisms of cellular therapy in respiratory diseases. Intensive Care Medicine. 2011;37(9):1421–1431.
    1. de Faria CA, de las Heras Kozma R, Stessuk T, Ribeiro-Paes JT. Experimental basis and new insights for cell therapy in chronic obstructive pulmonary disease. Stem Cell Reviews. 2012;8(4):1236–1244.
    1. Loi R, Beckett T, Goncz KK, Suratt BT, Weiss DJ. Limited restoration of cystic fibrosis lung epithelium in vivo with adult bone marrow-derived cells. American Journal of Respiratory and Critical Care Medicine. 2006;173(2):171–179.
    1. Bruscia EM, Ziegler EC, Price JE, Weiner S, Egan ME, Krause DS. Engraftment of donor-derived epithelial cells in multiple organs following bone marrow transplantation into newborn mice. Stem Cells. 2006;24(10):2299–2308.
    1. Bruscia EM, Price JE, Cheng E-C, et al. Assessment of cystic fibrosis transmembrane conductance regulator (CFTR) activity in CFTR-null mice after bone marrow transplantation. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(8):2965–2970.
    1. Agostini C. Stem cell therapy for chronic lung diseases: hope and reality. Respiratory Medicine. 2010;104(1):S86–S91.
    1. Ribeiro-Paes JT, Bilaqui A, Greco OT, et al. Unicentric study of cell therapy in chronic obstructive pulmonary disease/pulmonary emphysema. International Journal of COPD. 2011;6(1):63–71.
    1. Weiss DJ, Casaburi R, Flannery R, Leroux-Williams M, Tashkin DP. A placebo-controlled randomized trial of mesenchymal stem cells in chronic obstructive pulmonary disease. Chest. 2013;143(6):1590–1598.
    1. Spangrude GJ, Johnson GR. Resting and activated subsets of mouse multipotent hematopoietic stem cells. Proceedings of the National Academy of Sciences of the United States of America. 1990;87(19):7433–7437.
    1. Jones RJ, Collector MI, Barber JP, et al. Characterization of mouse lymphohematopoietic stem cells lacking spleen colony-forming activity. Blood. 1996;88(2):487–491.
    1. Jordan CT, Yamasaki G, Minamoto D. High-resolution cell cycle analysis of defined phenotypic subsets within primitive human hematopoietic cell populations. Experimental Hematology. 1996;24(11):1347–1355.
    1. Uchida N, Jerabek L, Weissman IL. Searching for hematopoietic stem cells. II. The heterogeneity of Thy-1.1(lo)Lin(-/lo)Sca-1+ mouse hematopoietic stem cells separated by counterflow centrifugal elutriation. Experimental Hematology. 1996;24(5):649–659.
    1. Goodell MA, Rosenzweig M, Kim H, et al. Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species. Nature Medicine. 1997;3(12):1337–1345.
    1. Gothot A, Pyatt R, McMahel J, Rice S, Srour EF. Functional heterogeneity of human CD34+ cells isolated in subcompartments of the G0/G1 phase of the cell cycle. Blood. 1997;90(11):4384–4393.
    1. Gothot A, Pyatt R, McMahel J, Rice S, Srour EF. Assessment of proliferative and colony-forming capacity after successive in vitro divisions of single human CD34+ cells initially isolated in G0. Experimental Hematology. 1998;26(7):562–570.
    1. Wiesmann A, Phillips RL, Mojica M, et al. Expression of CD27 on murine hematopoietic stem and progenitor cells. Immunity. 2000;12(2):193–199.
    1. Dykstra B, Kent D, Bowie M, et al. Long-term propagation of distinct hematopoietic differentiation programs in vivo. Cell Stem Cell. 2007;1(2):218–229.
    1. Kent DG, Copley MR, Benz C, et al. Prospective isolation and molecular characterization of hematopoietic stem cells with durable self-renewal potential. Blood. 2009;113(25):6342–6350.
    1. Sieburg HB, Cho RH, Dykstra B, Uchida N, Eaves CJ, Muller-Sieburg CE. The hematopoietic stem compartment consists of a limited number of discrete stem cell subsets. Blood. 2006;107(6):2311–2316.
    1. Benveniste P, Frelin C, Janmohamed S, et al. Intermediate-term hematopoietic stem cells with extended but time-limited reconstitution potential. Cell Stem Cell. 2010;6(1):48–58.
    1. Lemischka IR, Raulet RH, Mulligan RC. Developmental potential and dynamic behavior of hematopoietic stem cells. Cell. 1986;45(6):917–927.
    1. Challen GA, Boles NC, Chambers SM, Goodell MA. Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-β1. Cell Stem Cell. 2010;6(3):265–278.
    1. Schroeder T. Hematopoietic stem cell heterogeneity: subtypes, not unpredictable behavior. Cell Stem Cell. 2010;6(3):203–207.
    1. Cross MA, Heyworth CM, Murrell AM, Bockamp EO, Dexter TM, Green AR. Expression of lineage restricted transcription factors precedes lineage specific differentiation in a multipotent haemopoietic progenitor cell line. Oncogene. 1994;9(10):3013–3016.
    1. Chang HH, Hemberg M, Barahona M, Ingber DE, Huang S. Transcriptome-wide noise controls lineage choice in mammalian progenitor cells. Nature. 2008;453(7194):544–547.
    1. Graf T, Enver T. Forcing cells to change lineages. Nature. 2009;462(7273):587–594.
    1. Schroeder T. Long-term single-cell imaging of mammalian stem cells. Nature Methods. 2011;8(4):S30–S35.
    1. Hope K, Bhatia M. Clonal interrogation of stem cells. Nature Methods. 2011;8(4):S36–S40.
    1. Phinney DG. Biochemical heterogeneity of mesenchymal stem cell populations: clues to their therapeutic efficacy. Cell Cycle. 2007;6(23):2884–2889.
    1. Russell KC, Phinney DG, Lacey MR, Barrilleaux BL, Meyertholen KE, O’Connor KC. In vitro high-capacity assay to quantify the clonal heterogeneity in trilineage potential of mesenchymal stem cells reveals a complex hierarchy of lineage commitment. Stem Cells. 2010;28(4):788–798.
    1. Russell KC, Lacey MR, Gilliam JK, Tucker HA, Phinney DG, O’Connor KC. Clonal analysis of the proliferation potential of human bone marrow mesenchymal stem cells as a function of potency. Biotechnology and Bioengineering. 2011;108(11):2716–2726.
    1. Wong AP, Keating A, Lu W-Y, et al. Identification of a bone marrow-derived epithelial-like population capable of repopulating injured mouse airway epithelium. Journal of Clinical Investigation. 2009;119(2):336–348.
    1. Kucia M, Wysoczynski M, Ratajczak J, Ratajczak MZ. Identification of very small embryonic like (VSEL) stem cells in bone marrow. Cell and Tissue Research. 2008;331(1):125–134.
    1. Ratajczak J, Wysoczynski M, Zuba-Surma E, et al. Adult murine bone marrow-derived very small embryonic-like stem cells differentiate into the hematopoietic lineage after coculture over OP9 stromal cells. Experimental Hematology. 2011;39(2):225–237.
    1. Dominici M, Pritchard C, Garlits JE, Hofmann TJ, Persons DA, Horwitz EM. Hematopoietic cells and osteoblasts are derived from a common marrow progenitor after bone marrow transplantation. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(32):11761–11766.
    1. Olmsted-Davis EA, Gugala Z, Camargo F, et al. Primitive adult hematopoietic stem cells can function as osteoblast precursors. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(26):15877–15882.
    1. Kassmer SH, Bruscia EM, Zhang P-X, Krause DS. Nonhematopoietic cells are the primary source of bone marrow-derived lung epithelial cells. Stem Cells. 2012;30(3):491–499.
    1. Kassmer SH, Jin H, Zhang PX, et al. Very small embryonic-like stem cells from the murine bone marrow differentiate into epithelial cells of the lung. Stem Cells. 2013;31(12):2759–2766.
    1. Kucia M, Reca R, Campbell FR, et al. A population of very small embryonic-like (VSEL) CXCR4+ SSEA-1+Oct-4+ stem cells identified in adult bone marrow. Leukemia. 2006;20(5):857–869.
    1. Anjos-Afonso F, Bonnet D. Nonhematopoietic/endothelial SSEA-1+ cells define the most primitive progenitors in the adult murine bone marrow mesenchymal compartment. Blood. 2007;109(3):1298–1306.
    1. Piro D, Piccoli C, Guerra L, et al. Hematopoietic stem/progenitor cells express functional mitochondrial energy-dependent cystic fibrosis transmembrane conductance regulator. Stem Cells and Development. 2012;21(4):634–646.
    1. Akashi K, Traver D, Miyamoto T, Weissman IL. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature. 2000;404(6774):193–197.
    1. Kondo M, Weissman IL, Akashi K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell. 1997;91(5):661–672.
    1. Piccoli C, Ria R, Scrima R, et al. Characterization of mitochondrial and extra-mitochondrial oxygen consuming reactions in human hematopoietic stem cells: novel evidence of the occurrence of NAD(P)H oxidase activity. Journal of Biological Chemistry. 2005;280(28):26467–26476.
    1. Trotta T, Di Gioia S, Piro D, et al. Effect of acute lung injury on VLA-4 and CXCR4 expression in resident and circulating hematopoietic stem/progenitor cells. Respiration. 2013;85(3):252–264.
    1. Quesenberry PJ. The continuum model of marrow stem cell regulation. Current Opinion in Hematology. 2006;13(4):216–221.
    1. Colvin GA, Dooner MS, Dooner GJ, et al. Stem cell continuum: directed differentiation hotspots. Experimental Hematology. 2007;35(1):96–107.
    1. Dooner MS, Aliotta JM, Pimentel J, et al. Conversion potential of marrow cells into lung cells fluctuates with cytokine-induced cell cycle. Stem Cells and Development. 2008;17(2):207–219.
    1. Xu J, Mora A, Shim H, Stecenko A, Brigham KL, Rojas M. Role of the SDF-1/CXCR4 axis in the pathogenesis of lung injury and fibrosis. American Journal of Respiratory Cell and Molecular Biology. 2007;37(3):291–299.
    1. May LA, Kicic A, Rigby P, et al. Cells of epithelial lineage are present in blood, engraft the bronchial epithelium, and are increased in human lung transplantation. Journal of Heart and Lung Transplantation. 2009;28(6):550–557.
    1. Bruscia EM, Zhang P-X, Ferreira E, et al. Macrophages directly contribute to the exaggerated inflammatory response in cystic fibrosis transmembrane conductance regulator-/-mice. American Journal of Respiratory Cell and Molecular Biology. 2009;40(3):295–304.
    1. Bonini C, Ferrari G, Verzeletti S, et al. HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia. Science. 1997;276(5319):1719–1724.
    1. Mastaglio S, Stanghellini MTL, Bordignon C, Bondanza A, Ciceri F, Bonini C. Progress and prospects: graft-versus-host disease. Gene Therapy. 2010;17(11):1309–1317.
    1. Zhao F, Zhang YF, Liu YG, et al. Therapeutic effects of bone marrow-derived mesenchymal stem cells engraftment on bleomycin-induced lung injury in rats. Transplantation Proceedings. 2008;40(5):1700–1705.

Source: PubMed

3
Prenumerera