2,533
Views
6
CrossRef citations to date
0
Altmetric
Reviews

Lung regeneration using amniotic fluid mesenchymal stem cells

&
Pages 447-451 | Received 26 Feb 2017, Accepted 29 May 2017, Published online: 04 Jul 2017

Abstract

Respiratory diseases, such as chronic obstructive pulmonary disease (COPD), pulmonary hypertension and lung fibrosis, are yet a major challenge in the world and they result in irreversible structural lung damage. Lung transplantation as the only therapeutic option face some major challenges like graft rejection and cancer, arising as a result of immunosuppression. A low survival rate faced by lung transplantation patients is presently limited to approximately 5 years. Lungs shortage therefore calls for a mechanism that would increase the availability of suitable organs for transplantation. In this review, we give an update on the use of amniotic fluid mesenchymal stem cells (AFMSCs) as an optimal source for lungs scaffold re-cellularization, due to their limitless accessibility and possibility for proliferation and differentiation. Further studies will be required in tissue engineering (TE) and regenerative medicine (RM), especially shifting our focus towards AFMSCs as a cell source for this regeneration.

Introduction

Chronic and acute lung diseases pose an increasing socio-economic problem [Citation1]. The main cause of perinatal morbidity and mortality is preterm birth. It accounts for >85% of all death and complications of perinatal [Citation2]. Perinatal care improvement has raised survival of extremely premature newborns [Citation3]. These infants, nevertheless, are prone to long-term lung and brain injury [Citation3]. Respiratory diseases cause differ, but the resulting damage of organ are comparable fibrosis and scaring, chronic inflammation results in loss of functional lungs tissue [Citation4].

Recent TE and RM advancement has led to the synthesis of functional organs or tissues in vitro and their positive integration into human recipients without rejection [Citation5,Citation6]. TE aims at fabricating organs or tissues to replace injured or damage body parts. Major components of this strategy includes a biomaterials; as the scaffold upon which other components are built, tissue-specific cells and a suitable environment. An appropriate combination of these constituents can direct the TE of tissue construct in vitro and in addition promotes functional tissue substitutes development after implantation in vivo [Citation7].

Studies have revealed some potential for the regeneration of adult and foetal lung after injury [Citation8–11], failure of lung to regenerate frequently leads to chronic inflammation, disease progression and pathogenesis. Approaches to regenerate injured lungs, through resident lung stem cells activation [Citation12–15], or through bone marrow-derived stem cells grafting [Citation16–18], have shown some success but need to be further validated. A more direct strategy based on directing pluripotent stem cells differentiation towards different lung epithelial cell kinds is obviously promising. However, initial determinations to differentiate embryonic stem cells into those of the pulmonary cells showed inadequate achievement [Citation19–22]. Recently, investigations have taken a stepwise strategies based on imitating the steps of lung development and have accomplished more favourable results.

The lung organogenesis

The lung is derived from the foregut endoderm. A quite number of patterning process cause the gut tube separation and other endodermal organs including pancreas, liver, eventually resulting in the generation of the lung buds and trachea [Citation23,Citation24]. Active reciprocal signalling between nearby mesenchyme and developing multipotent distal tip epithelium are necessary for lung buds stereotypical branching morphogenesis, likewise early differentiation events resulting in different cell lineages. The proximal epithelium was firstly formed from proximal progenitors with the emergence of neuroendocrine, basal, ciliated and secretory cells lining the maturing epithelium ( and ) [Citation25]. As branching morphogenesis proceeds, bronchioles ultimately branch into alveoli or millions of terminal air sacs, where gaseous exchange took place after birth. The respiratory epithelium is mainly made of two cells before birth; for gaseous exchange-type-I alveolar epithelial cells, and for secreting surfactants-type-II cells needed to stimulate breathing at birth. Lung maturation accomplishment occurs post-natally. As it matures, the turnover of cell decreases and is very slow in the completely mature adult lung ().

Figure 1. The diagram depict human lung at embryonic and adult stages. During lung development (upper panel), the proximal progenitors expressing NKX2.1 + SOX2 + develops to produce the proximal cells lineages seen in the adult epithelium (lower panel). The distal tip progenitors represented as NKX2.1 + SOX9 + or NKX2.1 + FOXP2 + give rise to the stereotypical branching morphogenesis, and thereafter the respiratory epithelial cells. This image was adapted from [Citation25] with copyright permission.

Figure 1. The diagram depict human lung at embryonic and adult stages. During lung development (upper panel), the proximal progenitors expressing NKX2.1 + SOX2 + develops to produce the proximal cells lineages seen in the adult epithelium (lower panel). The distal tip progenitors represented as NKX2.1 + SOX9 + or NKX2.1 + FOXP2 + give rise to the stereotypical branching morphogenesis, and thereafter the respiratory epithelial cells. This image was adapted from [Citation25] with copyright permission.

Table 1. Formation of anterior foregot endoderm using several factors and cocktails.

Table 2. Stepwise formation of lungs progenitor cells.

Endodermal bronchial buds and subsequent airway branches grow into the mesenchyme surrounding the thoracic gut tube. Deficiencies or abnormalities in the branching of the respiratory tree are the basis of many forms of pulmonary hypoplasia. Studies over the past several decades have demonstrated that branching morphogenesis of the respiratory tree is regulated by reciprocal interaction between the endoderm and surrounding mesoderm. For example, when mesenchyme in the region of the bifurcating bronchial buds is replaced with mesenchyme from around the developing trachea, further branching is inhibited.

Conversely, replacement of tracheal mesenchyme with that from the region of the bifurcating bronchial buds stimulates ectopic tracheal budding and branching. Based on experiments such as these, components of the extracellular matrix and growth factors have both been implicated in the stimulation and inhibition of branching.

For example, Collagens types IV and V, Laminin, Fibronectin, and Tenascin-all components of the extracellular matrix-are thought to play either a permissive or a stimulatory role in branching of the bronchial buds. Likewise, the regulation of expression of receptors for these matrix components has also been implicated in control of branching morphogenesis.

Mesenchymal stem cells

There is limited knowledge on lung mesenchymal precursors. However, there is report that small populations of resident lung cells that expresses certain phenotypic characteristics of mesenchymal stem cells (MSCs) with progenitor capacity exist within the lung. For example, resident lung “side population” (SP) cells have been isolated, they possess both epithelial and mesenchymal potential [Citation26–28]. These SP cells have been found to be resident at all levels of the airway tree, exhibiting a relatively constant phenotype, regardless of their source of isolation [Citation28]. Even though, it has been confirmed that these SP cells are a source of adult lung MSCs [Citation27], the role of SP cells in endogenous lung repair is yet to be fully understood. In addition, McQualter et al. described an endogenous population of fibroblastic progenitor cells with clonogenic ability in adult lung, which are mostly representative of MSC lineages [Citation29].

Repopulating injured and defective lung

AF is regarded as a crucial tool for the diagnosis of foetus well-being in pregnancy because of the role they play in the development of foetus. However, AF can safely be isolated by amniocentesis before culturing. Several different types of cells arises from AF and recent studies are looking into their potential for the application of cell therapy. The potentials of multipotent and pluripotent cells obtained from AF has been investigated and in vitro differentiations into several types of cells have effectively been carried out. Human amnion epithelial cells (hAECs), AFSCs and placental mesenchymal stem cells (PMSCs), have demonstrated the ability to differentiate into various cell types osteogenic, adipogenic, endothelial, myogenic, neurogenic, pulmonary, hepatogenic, pancreatic and cardiac lineages. In addition, in vivo investigation is stressing the importance and mechanisms of AFMSCs for therapy, particularly for lung and kidney diseases. AF may represent a good precious source for safely and easily retrievable types of cell that may be employed for RM purposes [Citation30].

With amniotic fluid mesenchymal stem cells

A novel stem cell source have been obtained from AF, AFMSCs is a promising source for regenerative medicine. These types of cells are of multipotent origin, having the potentials of differentiating into each of the embryonic germ layer cells. Carraro et al. [Citation31] studied hAFSC potential to differentiate into pulmonary lineages and integrate into murine lung following injury. Results of their investigations showed that hAFSC plasticity to respond to various kinds of lung damage in different ways through alveolar versus bronchiolar epithelial cell lineage markers specific expression, depends on injury type to recipient lung.

With amnion epithelial cells

AEC is also a promising source of cell in disease treatment such as COPD, acute respiratory distress syndrome, pulmonary oedema, pulmonary fibrosis and pulmonary hypertension. The transplantation of AECs in animal model for lung disease decreased subsequent fibrosis and inflammation and eventually improves the function of the lung [Citation4]. hAECs administration to bleomycin-induced lung diseased mice leads to a downregulation of interleukin-6, transforming growth factor-β, proinflammatory cytokines tumour necrosis factor-α, and interferon-γ, α-smooth muscle actin expression, reduced pulmonary collagen deposition and inflammatory cell infiltrate. Murphy et al. [Citation32] lately showed that hAECs reacts with host macrophages to exercise their regenerative potentials, probably through macrophages induction into an alternatively activated phenotype. Recent investigations have induced hAECs for the expression of lung-specific proteins [Citation33] like the ion channel cystic fibrosis transmembrane conductance regulator, signifying a crucial use of hAECs for cystic fibrosis therapy. These significant investigations shows that hAECs is a potential tool for bioengineering and clinical applications of lung disease patients.

Conclusions

Stem cell research had a significant progress during the past years and had turn out to be one the most attractive aspect of modern biology. Thanks to several studies, stem cells hold a significant valuable possibility in supporting and protecting native tissues throughout inflammatory and ischaemic-induced damage. AFMSCs hold a great potential. This review proposes AFMSCs to be a better source of stem cell for lungs regeneration because it has the ability to differentiate to all forms of cells; however, this stem cell choice needs further laboratory investigations.

Disclosure statement

No potential conflict of interest was reported by the authors.

References

  • Chapman KR, Mannino DM, Soriano JB, et al. Epidemiology and costs of chronic obstructive pulmonary disease. Eur Respir J. 2006;27:188–207.
  • Goldenberg RL, Culhane JF, Iams JD, et al. Epidemiology and causes of preterm birth,. Lancet. 2008;371:75–84.
  • Doyle LW, Anderson PJ, Battin M, et al. Long term follow up of high risk children: who, why and how?. BMC Pediatr. 2014;14:279.
  • Murphy S, Lim R, Dickinson H, et al. Human amnion epithelial cells prevent bleomycin-induced lung injury and preserve lung function. Cell transplant. 2011;20:909–923.
  • Atala A, Bauer SB, Soker S, et al. Tissue-engineered autologous bladders for patients needing cystoplasty, Lancet. 2006;367:1241–1246.
  • Raya-Rivera A, Esquiliano DR, Yoo JJ, et al. Tissue-engineered autologous urethras for patients who need reconstruction: An observational study. Lancet. 2011;377:1175–1182.
  • Langer R, Vacanti JP. Tissue Engineering. Science. 1993;260:920–926.
  • Evans MJ, Cabral LJ, Stephens RJ, Freeman G. Renewal of alveolar epithelium in the rat following exposure to NO2. Am J Pathol. 1973;70:175–198.
  • Evans MJ, Shami SG, Cabral-Anderson LJ, Dekker NP. Role of nonciliated cells in renewal of the bronchial epithelium of rats exposed to NO2. Am J Pathol. 1986;123:126–133.
  • Peake JL, Reynolds SD, Stripp BR, et al. Alteration of pulmonary neuroendocrine cells during epithelial repair of naphthalene-induced airway injury. Am J Pathol. 2000;156:279–286.
  • Van Winkle LS, Buckpitt a. R, Nishio SJ, et al. Cellular response in naphthalene-induced Clara cell injury and bronchiolar epithelial repair in mice. Am J Physiol. 1995;269:L800–L818.
  • McQualter JL, Yuen K, Williams B, et al. Evidence of an epithelial stem/progenitor cell hierarchy in the adult mouse lung. Proc Natl Acad Sci USA. 2010;107:1414–1419.
  • Teisanu RM, Lagasse E, Whitesides JF, et al. Prospective isolation of bronchiolar stem cells based upon immunophenotypic and autofluorescence characteristics. Stem Cells. 2009;27:612–622.
  • Kim CF, Jackson EL, Woolfenden AE, et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell. 2005;121:823–835.
  • Giangreco A, Arwert EN, Rosewell IR, et al. Stem cells are dispensable for lung homeostasis but restore airways after injury. Proc Natl Acad Sci USA. 2009;106:9286–9291.
  • Wong A, Keating PA, Lu W, et al. Identification of a bone marrow-derived epithelial-like population capable of repopulating injured mouse airway epithelium. J Clin Invest. 2009;119:336–348.
  • Gomperts BN, Belperio JA, Rao PN, et al. Circulating progenitor epithelial cells traffic via CXCR4/CXCL12 in response to airway injury. J Immunol. 2006;176:1916–1927.
  • Macpherson H, Keir P, Webb S, et al. Bone marrow-derived SP cells can contribute to the respiratory tract of mice in vivo. J Cell Sci. 2005;118:2441–2450.
  • Rippon HJ, Ali NN, Polak JM, et al. Initial observations on the effect of medium composition on the differentiation of murine embryonic stem cells to alveolar type II cells. Cloning Stem Cells. 2004;6:49–56.
  • Van Vranken BE, Rippon HJ, Samadikuchaksaraei A, et al. The differentiation of distal lung epithelium from embryonic stem cells. Curr Protoc Stem Cell Biol. 2007;Chapter 1:Unit 1G.1
  • Samadikuchaksaraei A, Cohen S, Isaac K, et al. Derivation of distal airway epithelium from human embryonic stem cells. Tissue Eng. 2006;12:867–875.
  • Ali NN, Edgar AJ, Samadikuchaksaraei A, et al. Derivation of type II alveolar epithelial cells from murine embryonic stem cells. Tissue Eng. 2002;8:541–550.
  • Que J, Choi M, Ziel JW, et al. Morphogenesis of the trachea and esophagus: Current players and new roles for noggin and Bmps, Differentiation. 2006;74:422–437.
  • Morrisey EE, Hogan BLM. Preparing for the first breath: genetic and cellular mechanisms in lung development,. Developmental Cell. 2010;18:8–23.
  • Firth AL, Dargitz CT, Qualls SJ, et al. Generation of multiciliated cells in functional airway epithelia from human induced pluripotent stem cells.. Proc Natl Acad Sci USA. 2014;111:E1723–E1730.
  • Majka SM, Beutz MA, Hagen M, et al. Identification of novel resident pulmonary stem cells: form and function of the lung side population. Stem Cells. 2005;23:1073–1081.
  • Martin J, Helm K, Ruegg P, et al. Adult lung side population cells have mesenchymal stem cell potential. Cytotherapy. 2008;10:140–151.
  • Reynolds S, Shen DH, Reynolds P, et al. Molecular and functional properties of lung SP cells. Am J Physiol Lung Cell Mol Physiol. 2007;292:L972–L983.
  • McQualter JL, Brouard N, Williams B, et al. Endogenous fibroblastic progenitor cells in the adult mouse lung are highly enriched in the sca-1 positive cell fraction. Stem Cells. 2009;27:623–633.
  • Da Sacco S, De Filippo RE, Perin L. Amniotic fluid as a source of pluripotent and multipotent stem cells for organ regeneration. Curr Opin Organ Transplant. 2011;16:101–105.
  • Carraro G, Perin L, Sedrakyan S, et al. Human amniotic fluid stem cells can integrate and differentiate into epithelial lung lineages. Stem Cells. 2008;26:2902–2911.
  • Murphy SV, Shiyun SC, Tan JL, et al. Human amnion epithelial cells do not abrogate pulmonary fibrosis in mice with impaired macrophage function. Cell transplant. 2012;21:1477–1492.
  • Ilancheran S, Michalska A, Peh G, et al. Stem cells derived from human fetal membranes display multilineage differentiation potential. Biol Reprod. 2007;77:577–588.
  • Green MD, Chen A, Nostro M-C, et al. Generation of anterior foregut endoderm from human embryonic and induced pluripotent stem cells. Nat Biotechnol. 2011;29:267–272.
  • Longmire TA, Ikonomou L, Hawkins F, et al. Efficient derivation of purified lung and thyroid progenitors from embryonic stem cells. Cell Stem Cell. 2012;10:398–411.
  • Gadue P, Huber TL, Paddison PJ, et al. Wnt and TGF-beta signaling are required for the induction of an in vitro model of primitive streak formation using embryonic stem cells. Proc Natl Acad Sci USA. 2006;103:16806–16811.
  • Goss AM, Tian Y, Tsukiyama T, et al. Wnt2/2b and beta-catenin signaling are necessary and sufficient to specify lung progenitors in the foregut. Dev Cell. 2009;17:290–298.
  • Harris-Johnson KS, Domyan ET, Vezina CM, et al. beta-Catenin promotes respiratory progenitor identity in mouse foregut. Proc Natl Acad Sci USA. 2009;106:16287–16292.
  • Domyan ET, Ferretti E, Throckmorton K, et al. Signaling through BMP receptors promotes respiratory identity in the foregut via repression of Sox2. Development. 2011;138:971–981.
  • Sekine K, Ohuchi H, Fujiwara M, et al. Fgf10 is essential for limb and lung formation.. Nat Genet. 1999;21:138–141.
  • Serls A, Doherty E, Parvatiyar SP, et al. Different thresholds of fibroblast growth factors pattern the ventral foregut into liver and lung. Development. 2005;132:35–47.
  • Metzger RJ, Klein OD, Martin GR, et al. The branching programme of mouse lung development. Nature. 2008;453:745–750.
  • Tompkins DH, Besnard V, Lange AW, et al. Sox2 activates cell proliferation and differentiation in the respiratory epithelium. Am J Respir Cell Mol Biol. 2011;45:101–110.
  • Tompkins DH, Besnard V, Lange AW, et al. Sox2 is required for maintenance and differentiation of bronchiolar Clara, ciliated, and goblet cells. PLoS One. 2009;4:e8248.
  • Que J, Luo X, Schwartz RJ, et al. Multiple roles for Sox2 in the developing and adult mouse trachea. Development. 2009;136:1899–1907.
  • Rockich BE, Hrycaj SM, Shih HP, et al. Sox9 plays multiple roles in the lung epithelium during branching morphogenesis. Proc Natl Acad Sci USA. 2013;110:E4456–E4464.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.