Skip to main content

Endogenous and exogenous stem cells: a role in lung repair and use in airway tissue engineering and transplantation

Abstract

Rapid repair of the denuded alveolar surface after injury is a key to survival. The respiratory tract contains several sources of endogenous adult stem cells residing within the basal layer of the upper airways, within or near pulmonary neuroendocrine cell rests, at the bronchoalveolar junction, and within the alveolar epithelial surface, which contribute to the repair of the airway wall. Bone marrow-derived adult mesenchymal stem cells circulating in blood are also involved in tracheal regeneration. However, an organism is frequently incapable of repairing serious damage and defects of the respiratory tract resulting from acute trauma, lung cancers, and chronic pulmonary and airway diseases. Therefore, replacement of the tracheal tissue should be urgently considered. The shortage of donor trachea remains a major obstacle in tracheal transplantation. However, implementation of tissue engineering and stem cell therapy-based approaches helps to successfully solve this problem. To date, huge progress has been achieved in tracheal bioengineering. Several sources of stem cells have been used for transplantation and airway reconstitution in animal models with experimentally induced tracheal defects. Most tracheal tissue engineering approaches use biodegradable three-dimensional scaffolds, which are important for neotracheal formation by promoting cell attachment, cell redifferentiation, and production of the extracellular matrix. The advances in tracheal bioengineering recently resulted in successful transplantation of the world's first bioengineered trachea. Current trends in tracheal transplantation include the use of autologous cells, development of bioactive cell-free scaffolds capable of supporting activation and differentiation of host stem cells on the site of injury, with a future perspective of using human native sites as micro-niche for potentiation of the human body's site-specific response by sequential adding, boosting, permissive, and recruitment impulses.

Introduction

Transplantation of the airway and lung tissue is an accepted modality of treatment for end-stage lung disease. Since the early 1990 s, more than 26,000 lung transplants have been performed at centers worldwide [1]. The most common indications, for which lung transplantation is performed, include cases of respiratory failure such as chronic obstructive pulmonary disease, cystic fibrosis (mucoviscidosis), idiopathic pulmonary fibrosis, idiopathic pulmonary hypertension, alpha-1 antitrypsin deficiency, bronchiestasis, and sarcoidosis [2]. However, the availability of donor tissues and organs is constantly limited, which presents a serious bottleneck for widespread transplantation surgery. The generation of bioengineered lung and tracheal tissue transplants, with the help of regenerative medicine, is considered a very promising alternative to the classical transplantation of donor organ/tissue.

Over two years ago, a successful transplantation of the world's first bioengineered trachea to a young woman with end-stage bronchomalacia was performed [3]. A donor trachea was first carefully decellularized using a soft detergent that prevented degradation and solubilization of the collagenous matrix. Major histocompatibility antigens were also removed from the donor trachea to prevent a transplant rejection reaction. The decellularized trachea was then seeded with two types of pre-expanded and predifferentiated autologous cells; i.e. mesenchymal stem cell-derived cartilage-like cells and epithelial respiratory cells. Finally, the bioengineered organ was engrafted into the recipient's body to replace the left main bronchus. After surgery, the patient did not develop any signs of antigenicity and continues to live a near-normal life.

The first tissue-engineered organ transplantation was still based on a donor trachea. However, to date, a variety of bioengineered tubular tracheal matrices were developed as an alternative to the donor's airway. When selecting new biomaterials for trachea bioengineering, researchers should evaluate a wide range of biological properties of candidate material including toxicity, toxigenicity, biocompatibility, biodegradability, durability, cell adhesion characteristics, and ability to mimic the function of a native organ as much as possible.

The epithelial cells-extracellular matrix (ECM) interactions play a crucial role in healing airway injuries and repair of the airway epithelium. The secretion of a provisional ECM, the cell-ECM relationships through epithelial receptors, and the remodeling of the ECM by matrix metalloproteinases contribute not only to airway epithelial repair by modulating epithelial cell migration and proliferation, but also to the differentiation of repairing cells, leading to the complete restoration of the wounded epithelium [4]. Therefore, while developing a bioengineered model of the human bronchiole, tissue engineers should pay special attention to the fabrication of biologically active scaffolds and matrices capable of fulfilling natural properties of the airway ECM, for example, by maintaining and slowly releasing factors essential for proliferation and differentiation of a stem cell transplant [5].

Another issue of challenge in lung regenerative medicine is the choice of an appropriate cell source to reconstitute the lung airway. Naturally, residual pools of adult stem cells (SCs) located within the basal layer of the upper airways, within or near pulmonary neuroendocrine cell rests, at the bronchoalveolar junction, and within the alveolar epithelial surface, are responsible for lung regeneration and repair [6]. Endogenous progenitor cells are also involved in lung regeneration, contributing particularly to the rapid repair of the denuded alveolar surface after injury [7, 8]. However, the repair capacity of lungs declines with age, which is primarily due to the endogenous SC failure. Therefore, the exogenous stem/progenitor cells, such as embryonic stem cells (ESCs), bone marrow-or fat-derived mesenchymal stem cells (MSCs), and recently amniotic fluid stem/progenitor cells, could be considered as an alternate cell source for lung regeneration. The limitation of xenogenic or allogenic SCs is their potential immunogenicity for the recipient organism that requires implementation of immunosuppression to minimize the risk of graft rejection.

Special attention should also be paid to the delivery of implant cells to the recipient site and providing boosting and recruitment impulses for survival, expansion, and differentiation of the stem cell transplant. The molecular physiology of complex interactions between the host and engrafted cells is far to be precisely understood and therefore needs further efforts to maximize the regeneration rate. In this review, we characterize the important role of cell-cell interactions and ECM in airway epithelium repair; consider the resources of endogenous and exogenous stem/progenitor cells that have been used or have a potential to be applied in lung regeneration; and analyze current strategies in tracheal bioengineering and transplantation.

Endogenous and exogenous stem and progenitor cells for lung repair

The airway epithelium is subjected to a lifetime exposure by inhaled particles and pathogens that may lead to the development of a variety of infectious and inflammatory respiratory diseases such as chronic bronchitis, asthma, chronic obstructive pulmonary disease, and cystic fibrosis. These pathologies are typically associated with changes in the architecture of the airway walls, which could vary from the epithelial structure remodeling to complete denudation of the basement membrane. To restore its functions, the airway epithelium has to rapidly repair the injuries and regenerate its structure and integrity. The regeneration process is a complex phenomenon that quickly starts after the lesion occurs. Epithelial cells at the wound edge dedifferentiate, spread, and migrate to cover the denuded area [9]. After migration, epithelial cells in the repairing area start to proliferate. Finally, to restore a functional mucociliary epithelium at the injury site, the epithelium forms a transitory squamous metaplasia followed by progressive redifferentiation [10].

Due to the very large size (> 70 m2) and spatial restrictions of the alveolar surface in adult humans, a large number of cells must function as a "ready reserve" to repair the damaged alveolar surface [11]. The repair of injuries and the regeneration of the epithelial structure involve stem and progenitor cells. The mechanism of the regeneration of the airway epithelium was widely studied in rodent models of lung injury. After epithelial damage in mouse, the sites of actively proliferating cells observed near the glandular ducts were referred to as basal cells providing evidence of SC niches [12]. These cell populations are heterogeneous and comprise subpopulations capable of either multipotent or unipotent differentiation leading to the restoration of a completely differentiated airway epithelium [13].

In the human fetus, both basal and suprabasal cells were able to reconstitute a fully differentiated airway epithelium after engraftment in a humanized xenograft model in severe combined immunodeficiency (SCID) mice suggesting for a similar progenitor potential [14]. In adult human airway epithelium, only isolated basal cells are capable of restoring a fully functional airway epithelium, but the adult secretory cells lose their regeneration potential compared to the fetal secretory cells [15]. Thus, although both secretory and basal cells are able to proliferate, only basal cells are now suggested to represent the SC compartment of the airway epithelium in tracheas and bronchi.

In rodent bronchioles, two types of cells, Clara cells and neuroendocrine cells localized in neuroepithelial bodies possess the ability to proliferate in response to bronchiolar and alveolar damages [16]. Among those, only a subset of Clara secretory protein-expressing cells, which are reside in the airway neuroepithelial bodies and bronchoalveolar duct junctions are able to reconstitute the bronchiolar airway epithelium and hence can be considered as bronchiolar SCs [17]. A population of endothelial cells resistant to bronchiolar and alveolar damages and which is capable of giving rise, not only to Clara cells, but also to type I and type II alveolar cells in vitro was found at the bronchioalveolar duct junction [18]. Indeed, this observation suggests that both bronchiolar and alveolar rodent epithelia could possess SC features.

Despite the well-documented nature of stem/progenitor cells in rodent bronchiolar airway epithelium, it remains to be clarified in humans. Recently, the multi-way analyzes of multicellular spheroids (termed as bronchospheres) produced by mechanical and enzymatic digestion of the adult human lung tissue revealed the presence of mixed phenotype cells with type II alveolar and Clara cell features and high expression of SC regulatory genes, which was either weakly or not detectable in original tissues [19]. These findings provide the evidence that adult human bronchioli, similarly to rodent bronchiolar and alveolar epithelia, should harbor SCs. Interestingly, the bronchospheres also exhibited mesenchymal features and, after silencing the Slug gene that plays a key role in epithelial-mesenchymal transition processes, they lost the SC-specific gene expression profile and gained a differentiated bronchial/alveolar phenotype [20]. This suggests that the epithelial-mesenchymal transition process could be induced in a subset of airway cells after injury of the adult human lung tissue.

The endogenous peripheral airway smooth muscle progenitors appear to occur very early in lung development. The peripheral mesenchyme that expresses fibroblast growth factor 10 (Fgf10) serves as a progenitor cell population for peripheral airway smooth muscle [21]. As the airway grows outwards, Fgf10-expressing airway smooth muscle progenitor cells spread along the expanding peripheral airway. The mesenchymal vascular progenitors (hemangioblasts) occur at the very early stages of the lung embryogenesis. Under the stimulation of vascular engothelium growth factor, which is secreted mainly by the primitive epithelium, these hemangioblasts differentiate into a capillary network surrounding the bronchial, lobar, and segmental branches of the airway [22].

Since adult human airway epithelial SCs were only recently discovered and their cultivation is still challenging, the researchers consider other sources of exogenous pluripotent SCs for airway tissue engineering, such as ESCs and MSCs. ESCs possess a great pluripotency since they are able to generate a variety of cell lineages including airway progenitor cells [23]. However, the application of human embryonic SCs is now limited due to the obvious ethical problems.

Bone marrow-derived adult MSCs circulating in blood were shown to be able to support lung repair in mice [24, 25]. There were two populations of those progenitor epithelial cells expressing distinct surface markers. The first population had epithelial characteristics as shown by cytokeratin expression, but also hematopoietic characteristics as shown by CD45 expression [24]. Another population of MSCs was positive for the early epithelial marker cytokeratin 5 (CK5) and the chemokine receptor CXCR4 [25]. Administration of FGF7 (also known as a keratinocyte growth factor) to mouse recipients of tracheal transplants resulted in enhanced engraftment of the CK5-positive progenitors to the injured proximal airway epithelium suggesting the role of FGF7 in local resident progenitor epithelial cell repair through the mobilization of subsets of CK5-positive epithelial progenitors [26].

Bone marrow-derived populations of MSCs, such as bone marrow-derived chondrocytes, were widely co-cultivated with respiratory epithelial cells to reconstitute artificially fabricated trachea constructs based on synthetic [27], composite [28], or natural decellularized matrixes [3, 29]. A tissue-engineered trachea seeded with bone marrow-derived chondrocytes and airway epithelial cells was successfully implanted into a human recipient [3]. At present, autologous bone marrow-derived MSCs seem to present the most popular SC type used in laryngotracheal tissue engineering. Adipose-derived MSCs may be also regarded as potentially suitable for the tracheal repair, but so far there are only a few reports about their use in the improvement of airway defects. Suzuki et al. [30] used fat-derived MSCs as part of a bioengineered scaffold to improve tracheal defects in rats, observing a well-differentiated and neovasularized airway epithelium two weeks post-implantation. A tissue-engineered trachea derived from a framed collagen scaffold, gingival fibroblasts, and adipose-derived SCs, showed good regeneration properties when implanted into rats with tracheal defects [31].

Human amniotic fluid SCs (hAFSCs) and umbilical blood cord (UBC)-derived SCs are new cell resources for lung regeneration. Human umbilical cord blood is a promising source for human MSCs. Hematopoietic SCs are present in the blood of the umbilical cord during and shortly after delivery. These SCs are in the blood at the time of delivery because they move from the liver (where blood formation takes place during fetal life) to the bone marrow (where blood is made after birth). UBC-derived SCs are similar to SCs that reside in the bone marrow. Higher healing properties of those cells were demonstrated in rodents. When administered intratracheally, human UCB-derived MSCs successfully attenuated the hyperoxia-induced lung injury in neonatal rats [32] and reduced fibrosis in the bleomycin-induced mouse model of lung injury through the activation of production of matrix metalloproteinases (MMPs) and inhibition of the impaired collagen synthesis [33]. Recently, a successful clinical application of human UCB-derived SCs for treatment of systemic lupus erythematosus-induced diffuse alveolar hemorrhage was reported. The cells were infused into the blood of a 19-year-old girl that showed dramatic improvements in her clinical condition, oxygenation level, radiographic and hematological status very soon after transplantation of MSCs [34].

Similarly to ESCs, hAFSCs are multipotent and capable of differentiating into cell types that represent each embryonic germ layer, including cells of adipogenic, osteogenic, myogenic, endothelial, neuronal, and hepatic lineages [35]. However, compared to ESCs, hAFSCs have a great advantage because they are not tumorigenic and teratogenic. Experiments in mice with lung injuries showed an excellent regeneration potential for hAFSCs, which were able to integrate into the murine lung and differentiate into pulmonary lineages after injury [36].

In 2006, an attractive possibility of the direct reprogramming of somatic cells to an embryonic stem cell-like pluripotent state was shown [37]. The reprogramming requires the ectopic expression of four or even fewer factors (Oct 4, Sox2, Nanog, and Klf4) responsible for maintaining pluripotency [38]. To maintain a pluripotency, human induced pluripotent stem (iPS) cells were shown to utilize signaling mechanisms that are similar to those used by human ESCs [39]. The iPS cells present a key advantage over true ESCs since they do not require an embryo to be sacrificed and ultimately will allow the autologous transplantation of induced SCs to repair damaged tissues [40]. To date, a variety of human and murine terminally differentiated somatic cells were reported to be reprogrammed into iPS cells. While no direct applications to airway cells have yet been reported, it is likely that such applications will become possible in the near future.

Early-passage iPSCs retained a transient epigenetic memory of their somatic cells of origin, which manifests as differential gene expression and altered differentiation capacity [41, 42]. These observations could be exploited in potential therapeutic applications to enhance differentiation into desired cell lineages.

It should be noted that in vitro reprogramming of somatic cells to iPS cells occurs with extremely low frequency and slow kinetics, suggesting the existence of a barrier factor. Cell senescence modulated by the activation of several negative cell cycle regulators, such as p53 (encoded by Trp53), p21 (encoded by Cdkn1a), and INK4a/ARF (encoded by Cdkn2a/2b), was considered to play a major barrier role for reprogramming [43–45]. Before the reprogramming stage, a preliminary down-regulation of these factors resulted in a marked increase (up to 28%) in the efficiency of reprogramming [46–48]. For example, silencing of p53 significantly increased the reprogramming efficiency of human somatic cells, directed with administration of only two pluripotency factors, Oct4 and Sox2 [47]. These results suggest new routes to more efficient reprogramming, avoiding the use of oncogenes for inducing pruripotency, and maximizing yield of new promising cell sources for tissue engineering.

Epithelial cell-extracellular matrix interactions and their mimic by bioengineered scaffolds

The airway epithelium plays a key role in wound healing through the release of ECM proteins and remodeling of the secreted provisional ECM. The epithelial cells secrete a range of factors contributing to airway repair and regeneration (Figure 1). They include structural matrix proteins (collagens, laminin, fibronectin, fibrin, etc.) and molecules modulating cell migration (integrins), cell-cell and cell-substrate interactions (glycans, cell adhesion receptors), ECM remodeling (MMPs), cell proliferation and differentiation (FGF7, epidermal growth factor (EGF), connective tissue growth factor, FGFs, and their receptors) [4]. The epithelial cells and inflammatory cells of the airways also release inflammatory mediators such as transforming growth factor (TGF)-β1 and tumor necrosis factor-α that influence the production of matrix molecules [49].

Figure 1
figure 1

Extracellular matrix proteins and their interaction with each other and with cell surface matrix receptors. Decorin, fibromodulin, and types IX and XI collagen all interact with type II collagen and regulate collagen fiber assembly and structure. Types II and VI collagen bind matrix receptors (integrins). Type II collagen can also bind to anchorin CII, whereas fibronectin binds to integrins. A large proteoglycan aggregate forms when multiple aggrecan molecules bind to a long strand of hyaluronic acid, which, in turn, is anchored to the cell by CD44. Additional matrix proteins shown include thrombospondin and cartilage oligomeric protein (COMP).

Tissue engineering presents a promising technique to create a functional tracheal substitute that may overcome many difficulties that other tracheal substitutes could not. Most cartilage tissue engineering approaches use biodegradable three-dimensional (3D) scaffolds, which are important for neocartilage formation by promoting chondrocyte attachment, cell redifferentiation, and production of extracellular cartilage matrix [50]. In the development of bioactive engineered matrices, current strategies try to utilize natural properties of the airway ECM as much as possible. Those include the creation of hybrid hydrogel 3 D networks containing a cell-binding site for ligation of cell-surface integrin receptors and substrates for MMPs, proteases implicated in wound healing and tissue regeneration [51, 52] and fabrication of scaffolds with immobilized signaling molecules (FGF [53], TGF-β1[54], etc.) that slowly release upon transplantation to support the tissue repair.

In order to increase non-integrin-dependent cell adhesion features of the engineered scaffold, using fibrin/hyaluronic acid (HA) composite as a scaffold biomaterial was suggested. HA (hyaluronan) is natively found in the cartilage tissue. This glucosaminoglycan functions as a core ECM molecule for the binding of keratin sulfate and chondroitin sulfate in forming aggrecan [55] and contributes to several cellular processes like cell proliferation, morphogenesis, inflammation, and wound repair [56]. Compared to relatively inert poly(ethylene glycol) (PEG) hydrogels, fibrin/HA gels exhibited better properties for supporting the differentiation of MSCs into chondrocytes as shown by enhanced expression of cartilage-specific markers by MSCs seeded into the fibrin/HA scaffold [57]. In humans, HA-based scaffold Hyalograft C (Fidia Advanced Biopolymers, Abano Terme, Italy) has been successfully applied in the treatment of chronic lesions of the knee and articular defect repair [58]. To date, Hyalograft C has not been used for tracheal repair in humans. The application of this scaffold for tracheal regeneration in animal models produced inconsistent results. A tissue-engineered trachea, fabricated from the fibrin/HA gel and autologous chondrocytes, and then transplanted into rabbits, showed successful regeneration and functional restoration of ciliated epithelium at the operated site without graft rejection and inflammation [59]. However, in another study, if implanted intra-or paralaryngeally, Hyalograft C exhibited biocompatibility-related problems initiating a foreign-body reaction and cartilage degradation in rabbits [60].

To avoid problems with biocompatibility, the use of scaffold-free cartilage grafts was proposed. The grafts were recently evaluated in laboratory animals. Autologous chondrocytes were cultivated in a bioreactor to fabricate scaffold-free cartilage sheets and then used for laryngotracheal reconstruction in rabbits. The scaffold-free engineered cartilage was capable to support the formation of a well-vascularized, autologous neotrachea, with excellent mechanical properties compatible with the rabbit's native trachea [61]. The grafts showed no signs of degradation or inflammatory reaction and were covered with mucosal epithelium. However, they did show signs of mechanical failure at the implantation site [62]. Overall, scaffold-free engineered cartilage represents a promising, new approach in tracheal reconstitution; however, further efforts are required to optimize its mechanical properties and biomaterial durability.

Strategies to deliver cell graft and support its survival and tracheal healing

Pioneering works involved the engraftment of human airway epithelial cells into immunodeficient (SCID) mice resulted in the development of a well-differentiated and functional human epithelium [63, 64]. Yang et al. [65] developed an approach for cultivating and scaffold-free delivery of epithelial cell sheets directly to host tissues. The utility of human respiratory epithelial cells for clinical transplantation is limited because those cells grow slowly. After transduction with a lentivirus-based vector, the growth rate and regeneration properties of transduced human epithelial cells have been significantly improved [66]. However, due to their viral modification, the clinical implication of transduced epithelial cells is still restricted by biosafety requirements.

Primary respiratory epithelial cells co-cultivated with other primary cells, such as fibroblasts and chondrocytes, and seeded into the collagenous or related carrier, were shown to reconstitute the structure of the tracheal wall, forming a fully differentiated airway epithelium and basement membrane located below the epithelium [67, 68]. To date, a range of experimental approaches and protocols for the in vitro development of lung tissue constructs composed of primary lung cells has been developed. For example, the development of a tissue-engineered model of human bronchiole consisting of primary cells and designed to study mechanisms of airway remodeling and lung inflammation in asthma has been recently reported [69].

Exogenous stem/progenitor cells may be delivered into the lung either intravenously, intratracheally, or by direct injection. The immediate efficiency of exogenous cell arrival and trapping in the lung is very high. However, the rate of extravasation of implanted cells from the capillary into the injured tissue and eventual integration into lung cell lineages was very low (< 5%) [70]. Better results could be produced when stem/progenitor cells are cultured with other cells and seeded into the natural or artificial scaffold. Co-cultivation of epithelial progenitors with autologous costal chondrocytes, smooth muscle cells, and respiratory ciliated epithelium followed by propagation in the natural decellularized matrix (i.e. pig jejunal segment with its own vascular pedicle) resulted in the development of a functional vascularized trachea containing the extracellular cartilaginous matrix [71]. The experience with the engineered pig trachea was then used in engineering human trachea produced after dissemination of cartilage-like MSCs and epithelial respiratory cells in the decellularized donor trachea, and successful transplantation of this construct into the recipient woman [3].

New trends in tissue-engineered tracheal transplantation

Huge efforts in the development of cell therapy-based approaches, tissue engineering techniques and their careful evaluation in animal models of lung diseases and induced airway injury, have yielded the first successful clinical applications in lung repair. The need for the introduction of new, efficient, and promising technologies for regeneration of tracheal and other defects in the lung tissues will increase along with the constantly rising number of people suffering from respiratory troubles.

Recent advances in airway tissue engineering provide a good opportunity for the treatment of a wide range of lung defects. In addition to the respiratory failure cases mentioned above, SC-based therapies show great potential for new clinical applications against acute respiratory distress syndrome [72], asthma [73], and bronchopulmonary dysplasia [74]. At present, the therapeutic potential of SCs is intensively assessed in rodent models of these diseases, with the possibility of proceeding to clinical trials [75–78].

The current regenerative medicine is leading to a new paradigm in medicine biotransferring heterologous concepts to the onset of autologous technologies that could lead to tissue regeneration in vivo. The new concept also includes studying and reproducing biological properties of ECM that regulates tissue differentiation in at least three ways: (i) the biochemical composition of the matrix constituents; (ii) the 3D-organization (architecture); and (iii) the mechanical forces mediated to the cells by the matrix. The in vivo ECM constitutes the biopolymer, which potentially plays a permissive role for tissue differentiation. The practical consequence of this research is the development of cell-free scaffolds capable of supporting activation and differentiation of host SCs on the site of injury.

The presence of the cell-free matrix and denuded tracheal segments stimulates the tracheal repair by attracting resident stem and non-stem epithelial cells to dedifferentiate, proliferate, expand over the denuned surface, and redifferentiate again [11]. By conjugating a collagen vitrigel membrane to a collagen sponge, Tada et al. [79] developed a bipotential collagen scaffold capable of promoting host epithelial cell growth and mesenchymal cell infiltration after transplantation to an animal model with tracheal defects. The new cell-free scaffold was successfully tested in rats and then in dogs. In the dog, the scaffold larynx implant was covered with soft tissue on day 18 post-surgery, followed by complete regeneration of the canine mucosa [80]. Recently, clinical trials showed good regeneration properties of this artificial scaffold for the repair of tracheal defects. The tracheoplasty of four patients (three with thyroid cancer and one with subglottic stenosis) with resection of the trachea and subsequent suturation of the defects with a cell-free scaffold (Marlex mesh tube covered by collagen sponge) resulted in a well-epithelialized airway lumen without any obstruction two months post-surgery [81, 82].

The next step of the practical development of this new concept is the use of the human native site as a micro-niche in order to potentiate the human body's site-specific response by adding boosting, permissive, and recruitment impulses [83]. This technique is expected to be cost-and labor-effective since it assumes the avoidance of any in vitro cell replication, expansion, and differentiation. The new approach is a multi-step process that will include the development of strategies involving a sequential implementation (treatment) of factors required for rapid activation of endogenous SCs at the affected site, attraction of exogenous SCs from the host circulation system, control of the local release of inflammatory cytokines and hypoxia, cell differentiation toward the terminal stage, and ECM remodeling. This strategy requires a deep knowledge of molecular mechanisms, temporal-spatial signaling networks, and cell-cell interactions contributing to the tracheal repair, as well as a careful and strict real-time control of the regeneration process.

To stimulate the proliferation potential of local somatic and progenitor cells, the ectopic expression of pluripotency factors may be used. For example, Sox17 required for early endoderm formation is able to reinduce multipotent progenitor cell behavior in mature lung cells [84]. To promote further differentiation of stem/progenitor cells, it is necessary to down-regulate production of Sox2 (important for branching of airways) [85] and consider the application of tissue-specific growth factors such as EGF, FGF7, basic FGF [26, 86]. Erythropoietin could be used as a boosting factor due to its emerging role in the repair of both hematopoietic and non-hematopoietic tissues [87, 88].

A successful realization of the new concept would ultimately benefit achieving the end point of the developments in regenerative medicine, which considers organ regeneration rather than tissue repair.

Abbreviations

ARF:

alternate reading frame, an alternative reading frame product of CDKN2A locus

a tumor suppressor gene

CD45:

cluster of differentiation 45

CD45:

antigen

protein tyrosine receptor-type phosphatase C

CDKN:

cyclin-dependent kinase inhibitor

CK5:

cytokeratin 5

CXCR4:

CXC chemokine receptor 4

3D:

three-dimensional

ECM:

extracellular matrix

EGF:

epidermial growth factor

ESC:

embryonic stem cell

FGF:

fibroblast growth factor

HA:

hyaluronic acid

hyaluronate

hAFSC:

human amniotic fluid stem cell

INK4a:

cyclin-dependent kinase inhibitor 2A (melanoma, p16, inhibits CDK4)

iPS:

inducible pluripotent stem cell

Klf4:

Krueppel-like factor 4, a key transcription factor in maintaining pluripotency

MMP:

matrix metalloproteinase

MSC:

mesenchymal stem cell

Nanog:

a key transcription factor in maintaining pluripotency

Oct4:

Octamer-4, a homeodomain transcription factor maintaining pluripotency

SC:

stem cell

SCID:

severe combined immunodeficiency

Sox2:

SRY (sex determining region Y)-box 2, a transcription factor

TGF:

transforming growth factor

Trp53:

transformation-related protein 53

a tumor suppressor gene

UBC:

umbilical blood cord

References

  1. Stehlik J, Edwards LB, Kucheryavaya AY, Aurora P, Christie JD, Kirk R, Dobbels F, Rahmel AO, Hertz MI: The Registry of the International Society for Heart and Lung Transplantation: Twenty-seventh official adult heart transplant report--2010. J Heart Lung Transplant. 2010, 29: 1089-1103. 10.1016/j.healun.2010.08.007.

    Article  PubMed  Google Scholar 

  2. Moffatt-Bruce SD: Lung transplantation. [http://emedicine.medscape.com/article/429499-overview]

  3. Macchiarini P, Jungebluth P, Go T, Asnaghi MA, Rees LE, Cogan TA, Dodson A, Martorell J, Bellini S, Parnigotto PP, Dickinson SC, Hollander AP, Mantero S, Conconi MT, Birchall MA: Clinical transplantation of a tissue-engineered airway. Lancet. 2008, 372: 2023-2030. 10.1016/S0140-6736(08)61598-6.

    Article  PubMed  Google Scholar 

  4. Puchelle E, Zahm JM, Tournier JM, Coraux C: Airway epithelial repair, regeneration, and remodeling after injury in chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2006, 3: 726-733. 10.1513/pats.200605-126SF.

    Article  CAS  PubMed  Google Scholar 

  5. Princz MA, Sheardown H: Heparin-modified dendrimer cross-linked collagen matrices for the delivery of basic fibroblast growth factor (FGF-2). J Biomater Sci Polym Ed. 2008, 19: 1201-1218. 10.1163/156856208785540181.

    Article  CAS  PubMed  Google Scholar 

  6. Snyder JC, Teisanu RM, Stripp BR: Endogenous lung stem cells and contribution to disease. J Pathol. 2009, 217: 254-264. 10.1002/path.2473.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. Reddy R, Buckley S, Doerken M, Barsky L, Weinberg K, Anderson KD, Warburton D, Driscoll B: Isolation of a putative progenitor subpopulation of alveolar epithelial type 2 cells. Am J Physiol Lung Cell Mol Physiol. 2004, 286: L658-L667. 10.1152/ajplung.00159.2003.

    Article  CAS  PubMed  Google Scholar 

  8. Lee J, Reddy R, Barsky L, Weinberg K, Driscoll B: Contribution of proliferation and DNA damage repair to alveolar epithelial type 2 cell recovery from hyperoxia. Am J Physiol Lung Cell Mol Physiol. 2006, 290: L685-L694. 10.1152/ajplung.00020.2005.

    Article  CAS  PubMed  Google Scholar 

  9. Zahm JM, Kaplan H, Herard AL, Doriot F, Pierrot D, Somelette P, Puchelle E: Cell migration and proliferation during the in vitro wound repair of the respiratory epithelium. Cell Motil Cytoskeleton. 1997, 37: 33-43. 10.1002/(SICI)1097-0169(1997)37:1<33::AID-CM4>3.0.CO;2-I.

    Article  CAS  PubMed  Google Scholar 

  10. Herard AL, Zahm JM, Pierrot D, Hinnrasky J, Fuchey C, Puchelle E: Epithelial barrier integrity during in vitro wound repair of the airway epithelium. Am J Respir Cell Mol Biol. 1996, 15: 624-632.

    Article  CAS  PubMed  Google Scholar 

  11. Warburton D, Perin L, DeFilippo R, Bellusci S, Shi W, Driscoll B: Stem/progenitor cells in lung development, injury repair, and regeneration. Proc Am Thorac Soc. 2008, 5: 703-706. 10.1513/pats.200801-012AW.

    Article  PubMed Central  PubMed  Google Scholar 

  12. Borthwick DW, Shahbazian M, Todd Krantz Q, Dorin J, Randell SH: Evidence for stem-cell niches in the tracheal epithelium. Am J Respir Cell Mol Biol. 2001, 24: 662-670.

    Article  CAS  PubMed  Google Scholar 

  13. Hong KU, Reynolds SD, Watkins S, Fuchs E, Stripp BR: In vivo differentiation potential of tracheal basal cells: evidence for multipotent and unipotent subpopulations. Am J Physiol Lung Cell Mol Physiol. 2004, 286: L643-L649. 10.1152/ajplung.00155.2003.

    Article  CAS  PubMed  Google Scholar 

  14. Avril-Delplanque A, Casal I, Castillon N, Hinnrasky J, Puchelle E, Peault B: Aquaporin-3 expression in human fetal airway epithelial progenitor cells. Stem Cells. 2005, 23: 992-1001. 10.1634/stemcells.2004-0197.

    Article  CAS  PubMed  Google Scholar 

  15. Hajj R, Baranek T, Le Naour R, Lesimple P, Puchelle E, Coraux C: Basal cells of the human adult airway surface epithelium retain transit-amplifying cell properties. Stem Cells. 2007, 25: 139-148. 10.1634/stemcells.2006-0288.

    Article  CAS  PubMed  Google Scholar 

  16. Hong KU, Reynolds SD, Giangreco A, Hurley CM, Stripp BR: Clara cell secretory protein-expressing cells of the airway neuroepithelial body microenvironment include a label-retaining subset and are critical for epithelial renewal after progenitor cell depletion. Am J Respir Cell Mol Biol. 2001, 24: 671-681.

    Article  CAS  PubMed  Google Scholar 

  17. 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: 173-182.

    Article  PubMed Central  PubMed  Google Scholar 

  18. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S, Crowley D, Bronson RT, Jacks T: Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 2005, 121: 823-835. 10.1016/j.cell.2005.03.032.

    Article  CAS  PubMed  Google Scholar 

  19. Tesei A, Zoli W, Arienti C, Storci G, Granato AM, Pasquinelli G, Valente S, Orrico C, Rosetti M, Vannini I, Dubini A, Dell'Amore D, Amadori D, Bonafe M: Isolation of stem/progenitor cells from normal lung tissue of adult humans. Cell Prolif. 2009, 42: 298-308. 10.1111/j.1365-2184.2009.00594.x.

    Article  CAS  PubMed  Google Scholar 

  20. Medici D, Hay ED, Olsen BR: Snail and Slug promote epithelial-mesenchymal transition through beta-catenin-T-cell factor-4-dependent expression of transforming growth factor-beta3. Mol Biol Cell. 2008, 19: 4875-4887. 10.1091/mbc.E08-05-0506.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  21. Mailleux AA, Kelly R, Veltmaat JM, De Langhe SP, Zaffran S, Thiery JP, Bellusci S: Fgf10 expression identifies parabronchial smooth muscle cell progenitors and is required for their entry into the smooth muscle cell lineage. Development. 2005, 132: 2157-2166. 10.1242/dev.01795.

    Article  CAS  PubMed  Google Scholar 

  22. Del Moral PM, Sala FG, Tefft D, Shi W, Keshet E, Bellusci S, Warburton D: VEGF-A signaling through Flk-1 is a critical facilitator of early embryonic lung epithelial to endothelial crosstalk and branching morphogenesis. Dev Biol. 2006, 290: 177-188. 10.1016/j.ydbio.2005.11.022.

    Article  CAS  PubMed  Google Scholar 

  23. Nishimura Y, Hamazaki TS, Komazaki S, Kamimura S, Okochi H, Asashima M: Ciliated cells differentiated from mouse embryonic stem cells. Stem Cells. 2006, 24: 1381-1388. 10.1634/stemcells.2005-0464.

    Article  CAS  PubMed  Google Scholar 

  24. MacPherson H, Keir PA, Edwards CJ, Webb S, Dorin J: Following damage, the majority of bone marrow derived airway cells express an epithelial marker. Respir Res. 2006, 7: 145-10.1186/1465-9921-7-145.

    Article  PubMed Central  PubMed  Google Scholar 

  25. Gomperts BN, Belperio JA, Rao PN, Randell SH, Fishbein MC, Burdick MD, Strieter RM: Circulating progenitor epithelial cells traffic via CXCR4/CXCL12 in response to airway injury. J Immunol. 2006, 176: 1916-1927.

    Article  CAS  PubMed  Google Scholar 

  26. Gomperts BN, Belperio JA, Fishbein MC, Keane MP, Burdick MD, Strieter RM: Keratinocyte growth factor improves repair in the injured tracheal epithelium. Am J Respir Cell Mol Biol. 2007, 37: 48-56. 10.1165/rcmb.2006-0384OC.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  27. Liu L, Wu W, Tuo X, Geng W, Zhao J, Wei J, Yan X, Yang W, Li L, Chen F: Novel strategy to engineer trachea cartilage graft with marrow mesenchymal stem cell macroaggregate and hydrolyzable scaffold. Artif Organs. 2010, 34: 426-433. 10.1111/j.1525-1594.2009.00884.x.

    Article  PubMed  Google Scholar 

  28. Le Visage C, Dunham B, Flint P, Leong KW: Coculture of mesenchymal stem cells and respiratory epithelial cells to engineer a human composite respiratory mucosa. Tissue Eng. 2004, 10: 1426-1435.

    Article  CAS  PubMed  Google Scholar 

  29. Go T, Jungebluth P, Baiguero S, Asnaghi A, Martorell J, Ostertag H, Mantero S, Birchall M, Bader A, Macchiarini P: Both epithelial cells and mesenchymal stem cell-derived chondrocytes contribute to the survival of tissue-engineered airway transplants in pigs. J Thorac Cardiovasc Surg. 2010, 139: 437-443. 10.1016/j.jtcvs.2009.10.002.

    Article  CAS  PubMed  Google Scholar 

  30. Suzuki T, Kobayashi K, Tada Y, Suzuki Y, Wada I, Nakamura T, Omori K: Regeneration of the trachea using a bioengineered scaffold with adipose-derived stem cells. Ann Otol Rhinol Laryngol. 2008, 117: 453-463.

    Article  PubMed  Google Scholar 

  31. Kobayashi K, Suzuki T, Nomoto Y, Tada Y, Miyake M, Hazama A, Wada I, Nakamura T, Omori K: A tissue-engineered trachea derived from a framed collagen scaffold, gingival fibroblasts and adipose-derived stem cells. Biomaterials. 2010, 31: 4855-4863. 10.1016/j.biomaterials.2010.02.027.

    Article  CAS  PubMed  Google Scholar 

  32. Chang YS, Oh W, Choi SJ, Sung DK, Kim SY, Choi EY, Kang S, Jin HJ, Yang YS, Park WS: Human umbilical cord blood-derived mesenchymal stem cells attenuate hyperoxia-induced lung injury in neonatal rats. Cell Transplant. 2009, 18: 869-886. 10.3727/096368909X471189.

    Article  PubMed  Google Scholar 

  33. Moodley Y, Atienza D, Manuelpillai U, Samuel CS, Tchongue J, Ilancheran S, Boyd R, Trounson A: Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol. 2009, 175: 303-313. 10.2353/ajpath.2009.080629.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  34. Liang J, Gu F, Wang H, Hua B, Hou Y, Shi S, Lu L, Sun L: Mesenchymal stem cell transplantation for diffuse alveolar hemorrhage in SLE. Nat Rev Rheumatol. 2010, 6: 486-489. 10.1038/nrrheum.2010.80.

    Article  PubMed  Google Scholar 

  35. De Coppi P, Bartsch G, Siddiqui MM, Xu T, Santos CC, Perin L, Mostoslavsky G, Serre AC, Snyder EY, Yoo JJ, Furth ME, Soker S, Atala A: Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol. 2007, 25: 100-106. 10.1038/nbt1274.

    Article  CAS  PubMed  Google Scholar 

  36. Carraro G, Perin L, Sedrakyan S, Giuliani S, Tiozzo C, Lee J, Turcatel G, De Langhe SP, Driscoll B, Bellusci S, Minoo P, Atala A, De Filippo RE, Warburton D: Human amniotic fluid stem cells can integrate and differentiate into epithelial lung lineages. Stem Cells. 2008, 26: 2902-2911. 10.1634/stemcells.2008-0090.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Takahashi K, Yamanaka S: Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006, 126: 663-676. 10.1016/j.cell.2006.07.024.

    Article  CAS  PubMed  Google Scholar 

  38. Geoghegan E, Byrnes L: Mouse induced pluripotent stem cells. Int J Dev Biol. 2008, 52: 1015-1022. 10.1387/ijdb.082640eg.

    Article  CAS  PubMed  Google Scholar 

  39. Vallier L, Touboul T, Brown S, Cho C, Bilican B, Alexander M, Cedervall J, Chandran S, Ahrlund-Richter L, Weber A, Pedersen RA: Signaling pathways controlling pluripotency and early cell fate decisions of human induced pluripotent stem cells. Stem Cells. 2009, 27: 2655-2666. 10.1002/stem.199.

    Article  CAS  PubMed  Google Scholar 

  40. Park IH, Arora N, Huo H, Maherali N, Ahfeldt T, Shimamura A, Lensch MW, Cowan C, Hochedlinger K, Daley GQ: Disease-specific induced pluripotent stem cells. Cell. 2008, 134: 877-886. 10.1016/j.cell.2008.07.041.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  41. Polo JM, Liu S, Figueroa ME, Kulalert W, Eminli S, Tan KY, Apostolou E, Stadtfeld M, Li Y, Shioda T, Natesan S, Wagers AJ, Melnick A, Evans T, Hochedlinger K: Cell type of origin influences the molecular and functional properties of mouse induced pluripotent stem cells. Nat Biotechnol. 2010, 28: 848-855. 10.1038/nbt.1667.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  42. Kim K, Doi A, Wen B, Ng K, Zhao R, Cahan P, Kim J, Aryee MJ, Ji H, Ehrlich LI, Yabuuchi A, Takeuchi A, Cunniff KC, Hongguang H, McKinney-Freeman S, Naveiras O, Yoon TJ, Irizarry RA, Jung N, Seita J, Hanna J, Murakami P, Jaenisch R, Weissleder R, Orkin SH, Weissman IL, Feinberg AP, Daley GQ: Epigenetic memory in induced pluripotent stem cells. Nature.

  43. Banito A, Rashid ST, Acosta JC, Li S, Pereira CF, Geti I, Pinho S, Silva JC, Azuara V, Walsh M, Vallier L, Gil J: Senescence impairs successful reprogramming to pluripotent stem cells. Genes Dev. 2009, 23: 2134-2139. 10.1101/gad.1811609.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  44. Li H, Collado M, Villasante A, Strati K, Ortega S, Cañamero M, Blasco MA, Serrano M: The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature. 2009, 460: 1136-1139. 10.1038/nature08290.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  45. Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa M, Okita K, Yamanaka S: Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature. 2009, 460: 1132-1135. 10.1038/nature08235.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  46. Eminli S, Foudi A, Stadtfeld M, Maherali N, Ahfeldt T, Mostoslavsky G, Hock H, Hochedlinger K: Differentiation stage determines potential of hematopoietic cells for reprogramming into induced pluripotent stem cells. Nat Genet. 2009, 41: 968-976. 10.1038/ng.428.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  47. Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, Raya A, Wahl GM, Belmonte JC: Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature. 2009, 460: 1140-1144. 10.1038/nature08311.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  48. Utikal J, Polo JM, Stadtfeld M, Maherali N, Kulalert W, Walsh RM, Khalil A, Rheinwald JG, Hochedlinger K: Immortalization eliminates a roadblock during cellular reprogramming into iPS cells. Nature. 2009, 460: 1145-1148. 10.1038/nature08285.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  49. Lechapt-Zalcman E, Prulière-Escabasse V, Advenier D, Galiacy S, Charrière-Bertrand C, Coste A, Harf A, d'Ortho MP, Escudier E: Transforming growth factor-beta1 increases airway wound repair via MMP-2 upregulation: a new pathway for epithelial wound repair?. Am J Physiol Lung Cell Mol Physiol. 2006, 290: L1277-L1282. 10.1152/ajplung.00149.2005.

    Article  CAS  PubMed  Google Scholar 

  50. Raghunath J, Rollo J, Sales KM, Butler PE, Seifalian AM: Biomaterials and scaffold design: key to tissue-engineering cartilage. Biotechnol Appl Biochem. 2007, 46: 73-84. 10.1042/BA20060134.

    Article  CAS  PubMed  Google Scholar 

  51. Rizzi SC, Hubbell JA: Recombinant protein-co-PEG networks as cell-adhesive and proteolytically degradable hydrogel matrixes. Part I: Development and physicochemical characteristics. Biomacromolecules. 2005, 6: 1226-1238. 10.1021/bm049614c.

    Article  CAS  PubMed  Google Scholar 

  52. Zhu J: Bioactive modification of poly(ethylene glycol) hydrogels for tissue engineering. Biomaterials. 2010, 31: 4639-4656. 10.1016/j.biomaterials.2010.02.044.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  53. Komura M, Komura H, Kanamori Y, Tanaka Y, Suzuki K, Sugiyama M, Nakahara S, Kawashima H, Hatanaka A, Hoshi K, Ikada Y, Tabata Y, Iwanaka T: An animal model study for tissue-engineered trachea fabricated from a biodegradable scaffold using chondrocytes to augment repair of tracheal stenosis. J Pediatr Surg. 2008, 43: 2141-2146. 10.1016/j.jpedsurg.2008.08.038.

    Article  PubMed  Google Scholar 

  54. Jaklenec A, Hinckfuss A, Bilgen B, Ciombor DM, Aaron R, Mathiowitz E: Sequential release of bioactive IGF-I and TGF-beta 1 from PLGA microsphere-based scaffolds. Biomaterials. 2008, 29: 1518-1525. 10.1016/j.biomaterials.2007.12.004.

    Article  CAS  PubMed  Google Scholar 

  55. Morgelin M, Heinegard D, Engel J, Paulsson M: The cartilage proteoglycan aggregate: assembly through combined protein-carbohydrate and protein-protein interactions. Biophys Chem. 1994, 50: 113-128. 10.1016/0301-4622(94)85024-0.

    Article  CAS  PubMed  Google Scholar 

  56. Chen WY, Abatangelo G: Functions of hyaluronan in wound repair. Wound Repair Regen. 1999, 7: 79-89. 10.1046/j.1524-475X.1999.00079.x.

    Article  CAS  PubMed  Google Scholar 

  57. Chung C, Burdick JA: Influence of three-dimensional hyaluronic acid microenvironments on mesenchymal stem cell chondrogenesis. Tissue Eng Part A. 2009, 15: 243-254. 10.1089/ten.tea.2008.0067.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  58. Tognana E, Borrione A, De Luca C, Pavesio A: Hyalograft® C: Hyaluronan-based scaffolds in tissue-engineered cartilage. Cells Tissues Organs. 2007, 186: 97-103. 10.1159/000102539.

    Article  CAS  PubMed  Google Scholar 

  59. Kim DY, Pyun J, Choi JW, Kim JH, Lee JS, Shin HA, Kim HJ, Lee HN, Min BH, Cha HE, Kim CH: Tissue-engineered allograft tracheal cartilage using fibrin/hyaluronan composite gel and its in vivo implantation. Laryngoscope. 2010, 120: 30-38. 10.1002/lary.21108.

    Article  CAS  PubMed  Google Scholar 

  60. Weidenbecher M, Henderson JH, Tucker HM, Baskin JZ, Awadallah A, Dennis JE: Hyaluronan-based scaffolds to tissue-engineer cartilage implants for laryngotracheal reconstruction. Laryngoscope. 2007, 117: 1745-1749. 10.1097/MLG.0b013e31811434ae.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  61. Weidenbecher M, Tucker HM, Awadallah A, Dennis JE: Fabrication of a neotrachea using engineered cartilage. Laryngoscope. 2008, 118: 593-598. 10.1097/MLG.0b013e318161f9f8.

    Article  PubMed Central  PubMed  Google Scholar 

  62. Gilpin DA, Weidenbecher MS, Dennis JE: Scaffold-free tissue-engineered cartilage implants for laryngotracheal reconstruction. Laryngoscope. 2010, 120: 612-617. 10.1002/lary.20750.

    Article  CAS  PubMed  Google Scholar 

  63. Dupuit F, Gaillard D, Hinnrasky J, Mongodin E, de Bentzmann S, Copreni E, Puchelle E: Differentiated and functional human airway epithelium regeneration in tracheal xenografts. Am J Physiol Lung Cell Mol Physiol. 2000, 278: L165-L176.

    CAS  PubMed  Google Scholar 

  64. Puchelle E, Peault B: Human airway xenograft models of epithelial cell regeneration. Respir Res. 2000, 1: 125-128. 10.1186/rr21.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  65. Yang J, Yamato M, Nishida K, Ohki T, Kanzaki M, Sekine H, Shimizu T, Okano T: Cell delivery in regenerative medicine: the cell sheet engineering approach. J Control Release. 2006, 116: 193-203. 10.1016/j.jconrel.2006.06.022.

    Article  CAS  PubMed  Google Scholar 

  66. Castillon N, Avril-Delplanque A, Coraux C, Delenda C, Peault B, Danos O, Puchelle E: Regeneration of a well-differentiated human airway surface epithelium by spheroid and lentivirus vector-transduced airway cells. J Gene Med. 2004, 6: 846-856. 10.1002/jgm.570.

    Article  PubMed  Google Scholar 

  67. Nomoto Y, Suzuki T, Tada Y, Kobayashi K, Miyake M, Hazama A, Wada I, Kanemaru S, Nakamura T, Omori K: Tissue engineering for regeneration of the tracheal epithelium. Ann Otol Rhinol Laryngol. 2006, 115: 501-506.

    Article  PubMed  Google Scholar 

  68. Heikal MY, Aminuddin BS, Jeevanan J, Chen HC, Sharifah S, Ruszymah BH: A scanning electron microscopic study of in vivo tissue engineered respiratory epithelium in sheep. Med J Malaysia. 2008, 63 (Suppl A): 34-

    PubMed  Google Scholar 

  69. Miller C, George S, Niklason L: Developing a tissue-engineered model of the human bronchiole. J Tissue Eng Regen Med.

  70. Leblond AL, Naud P, Forest V, Gourden C, Sagan C, Romefort B, Mathieu E, Delorme B, Collin C, Pages JC, Sensebe L, Pitard B, Lemarchand P: Developing cell therapy techniques for respiratory disease: intratracheal delivery of genetically engineered stem cells in a murine model of airway injury. Hum Gene Ther. 2009, 20: 1329-1343. 10.1089/hum.2009.035.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  71. Walles T, Giere B, Hofmann M, Schanz J, Hofmann F, Mertsching H, Macchiarini P: Experimental generation of a tissue-engineered functional and vascularized trachea. J Thorac Cardiovasc Surg. 2004, 128: 900-906.

    Article  PubMed  Google Scholar 

  72. Lee JW, Gupta N, Serikov V, Matthay MA: Potential application of mesenchymal stem cells in acute lung injury. Expert Opin Biol Ther. 2009, 9: 1259-1270. 10.1517/14712590903213651.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  73. Iyer SS, Co C, Rojas M: Mesenchymal stem cells and inflammatory lung diseases. Panminerva Med. 2009, 51: 5-16.

    CAS  PubMed  Google Scholar 

  74. Bonfield TL, Caplan AI: Adult mesenchymal stem cells: an innovative therapeutic for lung diseases. Discov Med. 2010, 9: 337-345.

    PubMed  Google Scholar 

  75. Kumamoto M, Nishiwaki T, Matsuo N, Kimura H, Matsushima K: Minimally cultured bone marrow mesenchymal stem cells ameliorate fibrotic lung injury. Eur Respir J. 2009, 34: 740-748. 10.1183/09031936.00128508.

    Article  CAS  PubMed  Google Scholar 

  76. van Haaften T, Byrne R, Bonnet S, Rochefort GY, Akabutu J, Bouchentouf M, Rey-Parra GJ, Galipeau J, Haromy A, Eaton F, Chen M, Hashimoto K, Abley D, Korbutt G, Archer SL, Thébaud B: Airway delivery of mesenchymal stem cells prevents arrested alveolar growth in neonatal lung injury in rats. Am J Respir Crit Care Med. 2009, 180: 1131-1142. 10.1164/rccm.200902-0179OC.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  77. Nemeth K, Keane-Myers A, Brown JM, Metcalfe DD, Gorham JD, Bundoc VG, Hodges MG, Jelinek I, Madala S, Karpati S, Mezey E: Bone marrow stromal cells use TGF-beta to suppress allergic responses in a mouse model of ragweed-induced asthma. Proc Natl Acad Sci USA. 2010, 107: 5652-5657. 10.1073/pnas.0910720107.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  78. Wang D, Morales JE, Calame DG, Alcorn JL, Wetsel RA: Transplantation of human embryonic stem cell-derived alveolar epithelial type II cells abrogates acute lung injury in mice. Mol Ther. 2010, 18: 625-634. 10.1038/mt.2009.317.

    Article  PubMed Central  PubMed  Google Scholar 

  79. Tada Y, Suzuki T, Takezawa T, Nomoto Y, Kobayashi K, Nakamura T, Omori K: Regeneration of tracheal epithelium utilizing a novel bipotential collagen scaffold. Ann Otol Rhinol Laryngol. 2008, 117: 359-365.

    Article  PubMed  Google Scholar 

  80. Yamashita M, Omori K, Kanemaru S, Magrufov A, Tamura Y, Umeda H, Kishimoto M, Nakamura T, Ito J: Experimental regeneration of canine larynx: a trial with tissue engineering techniques. Acta Otolaryngol Suppl. 2007, 66-72. 10.1080/00016480601068014. 557

  81. Omori K, Nakamura T, Kanemaru S, Asato R, Yamashita M, Tanaka S, Magrufov A, Ito J, Shimizu Y: Regenerative medicine of the trachea: the first human case. Ann Otol Rhinol Laryngol. 2005, 114: 429-433.

    Article  PubMed  Google Scholar 

  82. Omori K, Tada Y, Suzuki T, Nomoto Y, Matsuzuka T, Kobayashi K, Nakamura T, Kanemaru S, Yamashita M, Asato R: Clinical application of in situ tissue engineering using a scaffolding technique for reconstruction of the larynx and trachea. Ann Otol Rhinol Laryngol. 2008, 117: 673-678.

    Article  PubMed  Google Scholar 

  83. Bader A, Macchiarini P: Moving towards in situ tracheal regeneration: the bionic tissue engineered transplantation approach. J Cell Mol Med. 2010, 14: 1877-1889. 10.1111/j.1582-4934.2010.01073.x.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  84. Lange AW, Keiser AR, Wells JM, Zorn AM, Whitsett JA: Sox17 promotes cell cycle progression and inhibits TGF-β/Smad3 signaling to initiate progenitor cell behavior in the respiratory epithelium. PLoS One. 2009, 4: e5711-10.1371/journal.pone.0005711.

    Article  PubMed Central  PubMed  Google Scholar 

  85. Gontan C, de Munck A, Vermeij M, Grosveld F, Tibboel D, Rottier R: Sox2 is important for two crucial processes in lung development: branching morphogenesis and epithelial cell differentiation. Dev Biol. 2008, 317: 296-309. 10.1016/j.ydbio.2008.02.035.

    Article  CAS  PubMed  Google Scholar 

  86. Lindsay CD: Novel therapeutic strategies for acute lung injury induced by lung damaging agents: The potential role of growth factors as treatment options. Hum Exp Toxicol.

  87. Arcasoy MO: The non-haematopoietic biological effects of erythropoietin. Br J Haematol. 2008, 141: 14-31. 10.1111/j.1365-2141.2008.07014.x.

    Article  CAS  PubMed  Google Scholar 

  88. Bader A, Machens HG: Recombinant human erythropoietin plays a pivotal role as a topical stem cell activator to reverse effects of damage to the skin in aging and trauma. Rejuvenation Res.

Download references

Acknowledgements

This work is supported by the grant 09-04-01420-a from the Russian Foundation for Basic Research

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Dimitry A Chistiakov.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution 2.0 International License (https://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Chistiakov, D.A. Endogenous and exogenous stem cells: a role in lung repair and use in airway tissue engineering and transplantation. J Biomed Sci 17, 92 (2010). https://doi.org/10.1186/1423-0127-17-92

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1423-0127-17-92

Keywords