1,507
Views
63
CrossRef citations to date
0
Altmetric
Special Focus Review

CD117+ amniotic fluid stem cells

State of the art and future perspectives

&
Pages 77-88 | Published online: 01 Jul 2012

Abstract

Broadly multipotent stem cells can be isolated from amniotic fluid by selection for the expression of the membrane stem cell factor receptor c-Kit, a common marker for multipotential stem cells. They have clonogenic capability and can be directed into a wide range of cell types representing the three primary embryonic lineages. Amniotic fluid stem cells maintained for over 250 population doublings retained long telomeres and a normal karyotype. Clonal human lines verified by retroviral marking were induced to differentiate into cell types representing each embryonic germ layer, including cells of adipogenic, osteogenic, myogenic, endothelial, neuronal and hepatic lineages. AFS cells could be differentiate toward cardiomyogenic lineages, when co-cultured with neonatal cardiomyocytes, and have the potential to generate myogenic and hematopoietic lineages both in vitro and in vivo. Very recently first trimester AFS cells could be reprogrammed without any genetic manipulation opening new possibilities in the field of fetal/neonatal therapy and disease modeling. In this review we are aiming to summarize the knowledge on amniotic fluid stem cells and highlight the most promising results.

Amniotic Fluid: Function, Origin and Composition

Amniotic fluid allows the fetus to freely grow and move inside the uterus, and acts as a vehicle for the exchange of body chemicals with the mother.Citation1,Citation2 In humans, the amniotic fluid starts to appear at the beginning of week 2 of gestation as a small film of liquid between the cells of the epiblast. Between days 8 and 10 after fertilization, this fluid gradually expands and separates the epiblast (i.e., the future embryo) from the amnioblasts (i.e., the future amnion), thus forming the amniotic cavity.Citation3 Thereafter, it progressively increases in volume, completely surrounding the embryo after week 4 of pregnancy. Over the course of gestation, amniotic fluid volume changes markedly from 20 ml in week 7 to 600 ml in week 25, 1,000 ml in week 34 and 800 ml at birth. During the first half of gestation, the amniotic fluid results from active sodium and chloride transport across the amniotic membrane and the non-keratinized fetal skin, with concomitant passive movement of water. In the second half of gestation, the amniotic fluid is constituted by fetal urine, gastrointestinal excretions, respiratory secretions and substances exchanged through the sac membranes.Citation4-Citation8 The amniotic fluid is primarily composed of water and electrolytes (98–99%) but also contains chemical substances (e.g., glucose, lipids, proteins, hormones and enzymes), suspended materials (e.g., vernix caseosa, lanugo hair and meconium) and cells. Amniotic fluid cells derive both from extra-embryonic structures (i.e., placenta and fetal membranes) and from embryonic and fetal tissues.Citation9 Although amniotic fluid cells are known to express markers of all three germ layers,Citation10,Citation11 their exact origin still represents a matter of discussion; the consensus is that they mainly consist of cells shed in the amniotic cavity from the developing skin, respiratory apparatus, urinary and gastrointestinal tracts.Citation6,Citation12-Citation14 Amniotic fluid cells display a broad range of morphologies and behaviors, varying with gestational age and fetal development. In normal conditions the number of amniotic fluid cells increases with advancing gestation; if a fetal disease is present, amniotic fluid cell counts can be either dramatically reduced (e.g., intrauterine death and urogenital atresia) or abnormally elevated (e.g., anencephaly, spina bifida and exomphalos).Citation14 Based on their morphological and growth characteristics, viable adherent cells from the amniotic fluid are classified into three main groups: epithelioid (33.7%), amniotic fluid (60.8%) and fibroblastic type (5.5%).Citation15 In the event of fetal abnormalities other types of cells can be found in the amniotic fluid, e.g., neural cells in presence of neural tube defects and peritoneal cells in case of abdominal wall malformations. The majority of cells present in the amniotic fluid are terminally differentiated and have limited proliferative capabilities.Citation16,Citation17 In the 1990s, however, two groups demonstrated the presence of small subsets of cells in the amniotic fluid harboring a proliferation and differentiation potential. First, Torricelli et al.Citation18 reported the presence of hematopoietic progenitors in the amniotic fluid collected before week 12 of gestation. Then Streubel et al.Citation19 were able to differentiate amniotic fluid cells into myocytes, thus suggesting the presence in the amniotic fluid of non-hematopoietic precursors. These results initiated a new interest in the amniotic fluid as an alternative source of cells for therapeutic applications.

CD117+ Amniotic Fluid Stem Cells

History

The first evidence that the amniotic fluid could contain pluripotent stem cells was provided when Prusa et al.Citation20 in 2003 described the presence of a distinct sub-population of proliferating amniotic fluid cells (0.1–0.5%) expressing the pluripotency marker Oct4 at both transcriptional and protein levels. Oct4 (i.e., octamer binding transcription factor 4) is a nuclear transcription factor that plays a critical role in maintaining ESC differentiation potential and capacity for self-renewal.Citation21,Citation22 Other than its expression by ESC, Oct4 is specifically expressed by germ cells, where its inactivation results in apoptosis, and by embryonal carcinoma cells and tumors of germ cell origin, where it acts as an oncogenic fate determinant.Citation23-Citation26 While its role in stem cells of fetal origin has not been completely addressed, it has been recently demonstrated that Oct4 is neither expressed nor required by somatic stem cells or progenitors.Citation27-Citation29 Despite this, different groups confirmed the expression of Oct4 and of its transcriptional targets (e.g., Rex-1) in the amniotic fluid.Citation30,Citation31 Remarkably, Karlmark et al.Citation32 transfected human amniotic fluid cells with the green fluorescent protein gene under either the Oct4 or the Rex-1 promoter and established that some amniotic fluid cells were able to activate these promoters. Several authors subsequently reported the possibility of harvesting amniotic fluid cells displaying features of pluripotent stem cells.Citation33,Citation34 Thereafter, the presence of a cell population able to generate clonal cell lines capable of differentiating into lineages representative of all three embryonic germ layers was definitively demonstrated.Citation35 These cells, named amniotic fluid stem cells (AFS cells), are characterized by the expression of the surface antigen c-kit (CD117), the type III tyrosine kinase receptor of the stem cell factor.Citation36

Isolation and culture

The proportion of c-kit positive/hematopoietic lineage negative cells in the amniotic fluid varies over the course of gestation roughly describing a Gaussian curve; they appear at very early time points in gestation (i.e., at 7 weeks of amenorrhea in humans and at E9.5 in mice) and present a peak at mid gestation corresponding to 90 × 104 cells/fetus at 20 weeks of pregnancy in humans and to 10,000 cells/fetus at E12.5 in mice ().Citation37 Human AFS cells have been derived either from small volumes (5 ml) of 1st (10–12 gestational weeks) and 2nd trimester AF (14–22 gestational weeks) or from confluent back-up amniocentesis cultures.Citation35,Citation38 Murine AFS cells have been obtained from the AF collected during the second week of gestation (E11.5−E15).Citation35,Citation37,Citation39 Rat AFS cells have been retrieved at a mean gestational age of 16 d.p.c.Citation40 AFS cell isolation is based on a two-step protocol consisting in the prior immunological selection (via flow cytometry or magnetic cell sorting) of CD117 positive cells from the AF (approximately 1% of total AF cells) and in the subsequent expansion of these cells in culture.Citation35,Citation37,Citation41-Citation43 Isolated AFS cells can be expanded in feeder layer-free, serum-rich conditions without evidence of spontaneous differentiation in vitro. Cells are cultured in basic medium containing 15% of fetal bovine serum and Chang supplement.Citation35,Citation37,Citation43 In a single study, murine AFS cells have been also successfully cultured onto feeder layers (i.e., mitomycin C-treated mouse embryonic fibroblasts) in basic medium supplemented with 15% fetal bovine serum, mercaptoethanol, stem cell factor (SCF), bone morphogenetic protein 4 (BMP4) and leukemia inhibitory factor (LIF).Citation44 Whether, these different culture protocols influence AFS cell features and potential remains to be established.

Figure 1. CD117 positive/hematopoietic lineage negative cells in the amniotic fluid of mice and human along the course of gestation. (A) Percentage of mouse AFS cells (mAFSC) as a function of gestational age. (B) Total number of mouse AFS cells (mAFSC) per embryo equivalent (EE) at different gestational ages. (C) Percentage of human AFS cells (hAFSC) as a function of gestational age. Means are represented by bars. Adapted from reference Citation37.

Figure 1. CD117 positive/hematopoietic lineage negative cells in the amniotic fluid of mice and human along the course of gestation. (A) Percentage of mouse AFS cells (mAFSC) as a function of gestational age. (B) Total number of mouse AFS cells (mAFSC) per embryo equivalent (EE) at different gestational ages. (C) Percentage of human AFS cells (hAFSC) as a function of gestational age. Means are represented by bars. Adapted from reference Citation37.

Characterization

Karyotype analysis of human AFS cells deriving from pregnancies in which the fetus was male revealed the fetal origin of these cells.Citation35,Citation38 AFS cells proliferate well during ex vivo expansion. When cultivated, they display a spectrum of morphologies ranging from a fibroblast-like to an oval-round shape (). AFS cells possess a great clonogenic potential.Citation35,Citation39,Citation42,Citation45 Clonal AFS cells lines expand rapidly in culture (doubling time = 36 h) and, more interestingly, maintain a constant telomere length (20 kbp) between early and late passages ().Citation35,Citation43 Despite their high proliferation rate, clonal AFS cells show a homogeneous, diploid DNA content without evidence of chromosomal rearrangement even after expansion to 250 population doublings (). Clonal AFS cells lines express markers of a pluripotent undifferentiated state: Oct4 and NANOG.Citation35,Citation39 However, it has been shown that they do not form tumors when injected undifferentiated into severe combined immunodeficient (SCID) mice.Citation35,Citation38,Citation46-Citation49 The cell-surface antigenic profile of AFS cells has been determined through flow cytometry by different investigators (). Cultured human AFS cells are positive for ESC (e.g., SSEA-4) and mesenchymal markers (e.g., CD73, CD90 and CD105), for several adhesion molecules (e.g., CD29 and CD44) and for antigens belonging to the major histocompatibility complex I (MHC-I). They are substantially negative for hematopoietic and endothelial markers (e.g., CD14, CD34, CD45, CD133 and CD31), and do not express antigens belonging to the major histocompatibility complex II (MHC-II), thus presenting a low immunogenicity profile.Citation35,Citation37,Citation47,Citation50,Citation51 As stability of cell lines is a fundamental prerequisite for basic and translational research, AFS cell capacity of maintaining their baseline characteristics over passages has been evaluated based on multiple parameters. Despite their high proliferation rate, AFS cells and derived clonal lines maintain constant morphology, doubling time, apoptosis rate, cell cycle distribution and marker expression (e.g., Oct4, CD117, CD29 and CD44) up to 25 passages.Citation43,Citation52 During in vitro expansion, however, cell volume tends to increase and significant fluctuations of proteins involved in different networks (i.e., signaling, antioxidant, proteasomal, cytoskeleton, connective tissue and chaperone proteins) can be observed using a gel-based proteomic approach.Citation52 The significance of these modifications warrants further investigations and needs to be taken in consideration.

Figure 2. (A) Human AFS cells mainly display a spindle-shaped morphology during in vitro cultivation under feeder layer-free, serum-rich conditions. (B and C) Clonal human AFS cell lines retain long telomeres and a normal karyotype after more than 250 cell divisions. (B) Conserved telomere length of AFS cells between early passage (20 population doublings, lane 3) and late passage (250 population doublings, lane 4). Short length (lane 1) and high length (lane 2) telomere standards provided in the assay kit. (C) Giemsa band karyogram showing chromosomes of late passage (250 population doublings) cells. Adapted from reference Citation35.

Figure 2. (A) Human AFS cells mainly display a spindle-shaped morphology during in vitro cultivation under feeder layer-free, serum-rich conditions. (B and C) Clonal human AFS cell lines retain long telomeres and a normal karyotype after more than 250 cell divisions. (B) Conserved telomere length of AFS cells between early passage (20 population doublings, lane 3) and late passage (250 population doublings, lane 4). Short length (lane 1) and high length (lane 2) telomere standards provided in the assay kit. (C) Giemsa band karyogram showing chromosomes of late passage (250 population doublings) cells. Adapted from reference Citation35.

Table 1. Surface markers expressed by human AFS cells: results by different groups

Potency

AFS cell broad multipotency has been established by demonstrating that clonal AFS cell lines: (1) can differentiate into cell lineages originating from all three primary germ layers under specific inducing conditions and (2) are able to form embryoid bodies (EB) when cultured in suspension. Starting from monoclonal lines, AFS cells (human, CD117-selected) could be differentiated in vitro along six distinct lineages representative of the three germ layers (adipogenic, osteogenic, myogenic, endothelial, neurogenic and hepatic) under specific culture conditions (). Cell clones were obtained by limited dilution and the single cell origin of clonal lines was confirmed via retroviral vector integration.Citation35 When cultured under conditions in which they are unable to attach to the surface of culture dishes and without anti-differentiation factors, embryonic stem (ES) cells harbor the potential to form three-dimensional aggregates, named EBs, which recapitulate the first steps of early mammalian embyogenesis.Citation53,Citation54 In 2010, for the first time, it was reported that starting from clonal human CD117-selected and AFS cell lines, embryoid bodies can be formed. The frequency of EB formation (i.e., % of number of EB recovered from 15 hanging drops) was estimated to be around 28% for AFS cell lines and around 67% for AFS cell clonal lines. Similarly to ES cells, EB generation by AFS cells is regulated by the mTor (i.e., mammalian target of rapamycin) pathway and is accompanied by a decreased expression of stem cell markers (e.g., nodal and Oct4) and by an increased expression of differentiation markers (e.g., nestin-ectodermal, GATA4-endodermal and Brachyury-mesodermal).Citation43,Citation55,Citation56 In 2012, it has been demonstrated that first trimester human CD117+ AFS cells represent a more undifferentiated and potent sub-population of AFS cells as they share 82% transcriptome identity with ES cells, express markers of pluripotency (OCT4, NANOG, SOX2 and c-MYC, KFL4) at a molecular and protein level and are able to form beating EBs with high efficiency. Within these EBs, AFS cells express markers representative of the three embryonic germ layers, with concomitant downregulation of OCT4.Citation38 Despite the aforementioned results, however, both 1st and 2nd trimester AFS cells cannot be rigorously considered pluripotent stem cells, as they do not form tumors when injected into nude mice and, so far, have not been shown to generate germline-competent chimeras.Citation35,Citation38,Citation39,Citation46-Citation49,Citation51,Citation57 Very recently, however, Moschidou et al.Citation38 have shown that human 1st trimester AFS cells can be reprogrammed to functional pluripotent stem cells by culture on Matrigel in ES cell medium supplemented with the histone deacetylase inhibitor (HDACi) valproic acid (VPA) but without ectopic reprogramming factors [i.e., amniotic fluid induced pluripotent stem (AFiPS) cells]. AFiPS cells show signs of true pluripotency as not only they differentiate into functional lineages of the three germ layers (bone, fat, cartilage, definitive endoderm, hepatocyte, ectoderm, neurons and oligo-dendrocytes) and readily form EBs in vitro, but also form teratomas after injection into immunocompromised mice. AFiPS cells express more than 200 genes in common with ES cells but uniquely express around 312 genes including those involved in spermatogenesis that are only active in germ cells. Complementary analysis revealed, in fact, that AFS cells share several common characteristics with primordial germ cells (PGC) including expression of CD117, T, FGF8, SOX17, STELLA, DAZL, NANOS, VASA, SSEA1, FRAGILIS and PUM2, thus suggesting that AFS cells might originate from PGC or PGC progenitors, which have been retained in the AF at the start of their migration to the genital ridge.Citation38 In vivo studies are needed to test the functional differentiation and tissue repair capacity of AFiPS cells in animal models of disease. However, these preliminary data suggest that AFiPS cells could provide an alternative source of pluripotent cells for allogeneic clinical use in cell-based therapies as they can be collected early in pregnancy during termination procedures and are not at potential risk of virally induced tumorigenicity as they are reprogrammed with VPA.

Figure 3. AFS cells differentiation. (A) Hepatogenic: urea secretion by human AFS cells before (rectangles) and after (diamonds) hepatogenic differentiation in vitro. (B) Neurogenic: secretion of neurotransmitter glutamic acid in response to potassium. (C) Osteogenic: mouse micro-CT scan 18 weeks after implantation of printed constructs of engineered bone from human AFS cells; arrow head: region of implantation of control scaffold without AFS cells; rhombus: scaffolds seeded with AFS cells. Adapted from reference Citation35.

Figure 3. AFS cells differentiation. (A) Hepatogenic: urea secretion by human AFS cells before (rectangles) and after (diamonds) hepatogenic differentiation in vitro. (B) Neurogenic: secretion of neurotransmitter glutamic acid in response to potassium. (C) Osteogenic: mouse micro-CT scan 18 weeks after implantation of printed constructs of engineered bone from human AFS cells; arrow head: region of implantation of control scaffold without AFS cells; rhombus: scaffolds seeded with AFS cells. Adapted from reference Citation35.

Preclinical studies

In the last ten years AFS cell research has become a fast-growing and promising area of interest. Many studies contributed to the today’s picture of the AF containing multipotent stem cells for different therapeutic applications. Hereafter, the most relevant papers investigating AFS cell therapeutic potential in the translational research setting are summarized; only studies explicitly employing CD117+ AFS cells have been considered.

Heart: in vitro differentiation toward the cardiac lineage and therapeutic potential in animal models of myocardial infarction

Cardiovascular diseases are the first cause of mortality in developed countries despite advances in pharmacological, interventional and surgical therapies. Cell transplantation is an attractive strategy to replace endogenous cardiomyocytes lost by myocardial infarction. Fetal and neonatal cardiomyocytes are the ideal cells for cardiac regeneration as they have been shown to integrate structurally and functionally into the myocardium after transplantation.Citation58 However, their application is limited by the ethical restrictions involved in the use of fetal and neonatal cardiac tissues.Citation59 Chiavegato et al.Citation47 were the first to investigate human AFS cell (human, 2nd trimester and CD117-selected) plasticity toward the cardiac lineage. Undifferentiated AFS cells express cardiac transcription factors at a molecular level (i.e., Nkx2.5 and GATA-4 mRNA) but do not produce any myocardial differentiation marker. The authors proved that, under in vitro cardiovascular inducing conditions, human AFS cells express cardiomyocyte (i.e., Nkx2.5, MLC-2v, GATA-4 and β-MyHC), endothelial (i.e., angiopoietin and CD146) and smooth muscle (i.e., smoothelin) markers. However, when xenotransplantated in a rat model of myocardial infarction, with or without cyclosporine treatment, or in intact heart from immuno-competent or immuno-deficient animals, AFS cells were acutely rejected and, in some animals with myocardial infarction, formed chondro-osteogenic masses.Citation59 Recently, Delo et al.Citation60 demonstrated that the chondro-osteogenic lesions reported by the latter study are independent from cell injection and related to infarction severity/size in the ischemic rat heart. AFS cell capacity of differentiating toward the cardiac lineage has been subsequently confirmed by several studies. Guan et al.Citation50 showed that undifferentiated AFS cells (human, 2nd trimester and CD117-selected): (1) express several cardiac genes including the transcription factor mef2, the gap junction connexin43 and H- and N-cadherin, (2) acquire a cardiomyocyte-like phenotype (i.e., morphological changes, upregulation of cardiac-specific genes troponin I and troponin T, redistribution of connexin43) after a 24-h incubation with 5-aza-2′-deoxycytidine (5-AZA-dC) and (3) form mechanical and electrical connections with neonatal rat cardiomyocytes when cultured together. Bollini et al.Citation40 proved that the co-colture with neonatal rat cardiomyocytes activates in AFS cells (rat, CD117-selected) a “myocardial gene program:” expression of myocardial antigenic markers (cTnI, MyHC and α-actinin) and development of electrical excitability and activity. In vivo, AFS cells (rat, CD117-selected) directly transplanted in an undifferentiated state in the heart of rats with ischemia/reperfusion injury (i.e., ligation of the left anterior descending coronary artery) engraft in the myocardium, differentiate into endothelial and smooth muscle cells and improve heart ejection fraction as measured 3 weeks after injury by MRI.Citation40 As proven in a similar model of acute myocardial infarction, AFS cell (human, 2nd trimester and CD117-selected) beneficial effects on infarct size and myocardial cell survival are exerted rapidly (few hours) by the secretion of paracrine effectors such as the cardioprotective and proangiogenic factor thymosin β4 (Tβ4).Citation61 AFS cells display a cardiogenic potential in vitro and in vivo; their potential use in the autologous setting (e.g., tissue engineering approaches to treat congenital cardiac malformations) as in the allogeneic context (e.g., ischemic injury) needs further evaluation.

Skeletal muscle: myogenic differentiation in vitro and muscle regeneration in animal models of disease

Stem cell therapy is an attractive method to treat muscular degenerative diseases because only a small number of cells, together with a stimulatory signal for expansion, are required to obtain a therapeutic effect.Citation62 The identification of a stem cell population providing efficient muscle regeneration is critical for the progression of cell therapy for muscle diseases.Citation63 Despite early data reporting AFS cell capacity of expressing molecular markers of muscle differentiation under specific culture conditions,Citation35 high impact papers have just been very recently published. First, Gekas et al.Citation48 demonstrated that human, 2nd trimester, CD117+-selected AFS cells can acquire a myogenic-like phenotype (i.e., expression of desmin and MyoD) after culture in myogenic-specific induction conditions (i.e., culture on plastic plates coated with Matrigel and 3 μM 5-aza-2′-deoxycytidine). However, when these cells were transplanted undifferentiated into uninjured muscles of SCID mice, they failed to differentiate toward the myogenic lineage in vivo. In 2012, Ma et al.Citation49 demonstrated for the first time that human, 2nd trimester, clonal AFS cell lines expressing (not selected for) CD117+ could differentiate into skeletal myogenic cells in vitro and incorporate into regenerating skeletal muscle in vivo. In vitro, after temporary exposure to the DNA demethylation agent 5-aza-2′-deoxycytidine or by co-culture with C2C12 myoblasts, AFS cells differentiated into skeletal myogenic cells expressing specific markers such as Desmin, Troponin I and α-Actinin. In vivo, four weeks after transplantation into cardiotoxin-injured and X-ray-irradiated muscles of NOD/SCID mice, AFS cells survived, differentiated into myogenic precursor cells (i.e., Desmin, Laminin and Myf5 expression) and fused with host myofibers, thus contributing to skeletal muscle regeneration. The authors hypothesized that undifferentiated AFS cells first migrated into the regenerating myofibers driven by cytokines or chemokines, then converted into myogenic precursors residing in the satellite cell niche as demonstrated by Myf-5 expression, and finally were activated to fuse with the myofibers. Very recently, Piccoli et al.Citation44 demonstrated for the first time the functional and stable long-term integration of AFS cells into the skeletal muscle of HSA-Cre SmnF7/F7 mutant mice, which closely replicate the clinical features of human muscular dystrophies. AFS cells were obtained from E11.5–13.5 GFP+ mice through immediate CD117 selection after amniotic fluid collection. Approximately 25,000 freshly isolated AFS cells were directly injected into the tail vein of each animal without previous expansion in culture. Transplanted mice displayed enhanced muscle strength, improved survival rate by 75% and restored muscle phenotype in comparison to untreated animals. Not only, dystrophin distribution in GFP+ myofibers was similar to that of wild type animals, but also GFP+ cells were found engrafted into the muscle stem cell niche as demonstrated by their sub-laminal position and by Pax7 and α7integrin expression. Functional integration of AFS cells in the stem cell niche was confirmed by successful secondary transplants of GFP+ satellite cells derived from AFS cell-treated mice into untreated SmnF7/F7 mutant mice. In order to progress toward their application for therapy, the therapeutic potential of cultivated AFS cells was also investigated and 25,000 AFS cells previously expanded in culture were intravenously injected into SmnF7/F7 mice. Despite maintaining a therapeutic potential, cultivated AFS cells regenerated approximately 20% of the recipient muscle fibers vs. 50% when employing freshly isolated AFS cells, thus, highlighting the importance of optimizing AFS cell expansion conditions. In summary, available data support that AFS cells have the potential: (1) to differentiate toward the myogenic lineage, (2) to participate into muscle regeneration in animal models of muscle injury and (3) to engraft and to participate into the muscle stem cell niche. Hence, AFS cells do constitute a promising therapeutic option for muscle skeletal muscle degenerative diseases. Understanding the process of AFS cell myogenesis in vivo and improving cell expansion in vitro will be crucial requisites before this could happen.

Bone: differentiation in vitro and bone repair in vivo

Critical-sized segmental bone defects are one of the most challenging problems faced by orthopedic surgeons. Autologous and heterologous bone grafting are limited respectively by the small amount of tissue available for transplantation and by high re-fracture rates.Citation64,Citation65 Tissue engineering strategies that combine biodegradable scaffolds with stem cells capable of osteogenesis have been indicated as promising alternatives to bone grafting;Citation66 however, bone regeneration through cell-based therapies has been limited so far by the insufficient availability of osteogenic cells.Citation67 The potential of AFS cells to synthesize in vitro mineralized extracellular matrix within porous scaffolds has been investigated by different groups. After exposure to osteogenic conditions in static 2D cultures, AFS cells differentiate into functional osteoblasts (i.e., activate the expression of osteogenic genes such as Runx2, Osx, Bsp, Opn, Ocn and produce alkaline phosphatase) and form dense layers of mineralized matrix.Citation35,Citation67-Citation70 When seeded into 3D biodegradable scaffolds and stimulated by osteogenic supplements (i.e., rhBMP-7 or dexamethasone), AFS cells show an even more significant osteogenic differentiation and production of mineralized matrix in comparison to 2D cultures.Citation35,Citation67-Citation70 In vivo, AFS cells are not able to mineralize at ectopic sites unless previously pre-differentiated in vitro.Citation67 However, when seeded into 3D scaffolds and pre-commited in vitro toward the osteogenic lineage, AFS cells are able to repair critical sized bone defects. Rodrigues et al.Citation71 initially deposited AFS cells (human, 2nd trimester and CD117-selected) into starch-poly(ε-caprolactone) (SPCL) scaffolds and cultured them for three weeks in osteogenic conditions; then implanted the constructs composed by AFS cells plus SPCL scaffolds into critical sized femoral defects of rats. In vivo formation of new bone was observed by micro-CT imaging with complete repair of bone defect after 16 weeks after implantation. Similarly, pre-differentiated AFS cells (human, 2nd trimester and CD117-selected) seeded into silk fibroin scaffolds were able to repair critical-size cranial bone defects in immunocompromised rats via formation mature bone in a four week time.Citation72 These results demonstrate the potential of AFS cells to produce 3D mineralized bioengineered constructs and suggest that AFS cells may be an effective cell source for the autologous and heterologous repair of large bone defects. Further studies are needed to confirm the long-term functional performance of these grafts into bone injury sites.

Cartilage: in vitro differentiation into functional chondrocytes

Enhancing the regeneration potential of hyaline cartilage is one of the most significant challenges for treating damaged cartilage.Citation73 The capacity of AFS cells to differentiate into chondrocytes has been tested in vitro. Human CD117-selected AFS cells treated with TGF-beta1 have been proven to produce significant amounts of cartilaginous matrix (i.e., sulfated glycosaminoglycans and type II collagen) both in pellet and alginate hydrogel cultures.Citation74 Via cultivation of AFS cells (human and CD117-selected) into a defined osteochondral medium, Rodrigues et al.Citation71 generated bilayered osteochondral constructs consisting, on one side, of a SPCL scaffold containing AFS cells differentiated toward the osteogenic lineage and, on the other side, of a agarose hydrogel seeded with AFS cells committed toward the chondrogenic lineage. The regenerative potential of pre-differentiated cells and engineered constructs should be tested in vivo.

Kidney: AFS cell differentiation in organotypic cultures and therapeutic potential in animal models of acute and chronic disease

The incidence and prevalence of end stage renal disease (ESRD) continues to increase worldwide. Although renal transplantation represents a good treatment option, the shortage of compatible organs remains a critical issue for patients affected by ESRD. Therefore, the possibility of developing stem cell-based therapies for both glomerular and tubular repair is receiving intensive investigation.Citation75 Different stem cell types have shown some potential in the generation of functional nephronsCitation76-Citation81 but the most appropriate cell type for transplantation is still to be established.Citation82 The first evidence that AFS cells could be of relevance for renal stem cell therapy has been published in 2007. Perin et al.Citation42 demonstrated that monoclonal CD117+ human 2nd trimester AFS cells: (1) do not basally express kidney markers and thus are not specifically pre-committed to kidney progenitor cells and (2) if injected into isolated murine embryonic kidneys can integrate into renal tissue, participate in all steps of nephrogenesis and express molecular markers of kidney differentiation such as ZO-1, GDNF and claudin. In 2010, Siegel et al.Citation45 definitely proved that monoclonal human CD117-selected AFS cells could contribute to kidney development. Chimeric organotypic renal structures were generated via mixing single-cell suspensions of murine embryonic kidneys with human AFS cells. Descending from one cell, pluripotent Oct4- and CD117-positive AFS cells were able to contribute to renal tissue formation and to express renal specific markers (e.g., Pax2-ureteric bud and nephron structures marker, E-cadherin-distal tubule marker, calbindin-ureteric bud marker and laminin-marker of epithelial structures in the developing kidney). Moreover, it was proven that AFS cell contribution to nephrogenesis is regulated by components of the mammalian target of rapamycin (mTOR) signaling pathway (i.e., mTORC1 and mTORC2).Citation45 The first successful therapeutic application of AFS cells in an animal model of renal disease was published in 2010. Clonal cell lines derived from CD117+ human 2nd trimester AFS cells were directly injected into the kidneys of nude mice affected by acute tubular necrosis (ATN) due to glycerol-induced rhabdomyolysis. When administered at the time of damage, AFS cells provided a strong protective effect toward ATN injury, as reflected by the more rapid resolution of tubular structural damage, decreased tubular cell apoptosis, increased tubular cell proliferation and reduced creatinine and BUN levels. Despite these striking results, the efficiency of AFS cell integration into the renal tissue was low and just few integrated cells showed a commitment toward renal differentiation. Multiplex cytokine assays demonstrated a lower expression of pro-inflammatory molecules (e.g., IL16 and IL27) and a higher expression of anti-inflammatory cytokines (e.g., IL10, IL1Ra and IL6) in the kidneys of mice treated with AFS cells vs. placebo, thus suggesting an immune-modulatory effect of AFS cells in the prevention/resolution of renal damage during ATN.Citation57 Comparable results have been obtained by Hauser et al.,Citation83 who injected a heterogenous mixture of 2nd trimester human amniotic fluid-derived cells (non CD117-selected and non-clonal) into nude mice with glycerol-induced acute kidney injury (AKI) obtaining normalization of renal function. Similarly to the previous study, the injected cells were found to persistently accumulate within the peritubular capillaries and the interstitium and to induce enhanced tubular cell proliferation and reduced apoptosis in a differentiation-independent mechanism. However, as a heterogenous cell population was employed, the authors could not exclude the employment of renal-progenitor cells (known to be contained in human amniotic fluid) and could not guarantee the utilization of a homogeneous population of stem cells. Another proof of AFS cell beneficial effect in AKI models was recently given by Rota et al.Citation51 Infusion of human, 2nd trimester, CD117+ AFS cells into NOD-SCID mice with cisplatin-induced AKI improved renal function, limited tubular damage and prolonged animal survival. AFS cells predominantly engrafted in the peritubular regions without showing signs of tubular differentiation. They exerted anti-apoptotic effect and stimulated proliferation of tubular cells via local release of proteic factors, including IL6, VEGF and SDF1/CXCL12. The therapeutic potential of AFS cells was enhanced by cell pretreatment with glial cell line-derived neurotrophic factor (GDNF), which markedly ameliorated renal function and tubular injury by increasing AFS cell production of growth factors, motility, and expression of receptors involved in cell homing and survival.Citation51 After proving the beneficial role of AFS cells in acute kidney damage (i.e., ATN), Perin’s group has also recently demonstrated AFS cell therapeutic potential in a model of chronic kidney disease. CD117-selected murine clonal AFS cell lines were systemically injected into genetically modified mice affected by Alport Syndrome (Col4a5−/−) during the very early phase of disease (i.e., before onset of proteinuria). A single injection of AFS cells delayed interstitial fibrosis and progression of glomerular sclerosis, prolonged animal survival, and ameliorated the decline of kidney function. AFS cells did not differentiate into podocyte-like cells and did not stimulate production of the collagen IVa5 needed for normal formation and function of the glomerular basement membrane. Instead, the mechanism of renal protection consisted in the paracrine/endocrine modulation of pro-fibrotic cytokine expression and of macrophages recruitment to the interstitial space.Citation39 Taken together, the above reported studies provide evidence that AFS cells harbor the potential to differentiate upon renal lineages and warrant further investigation of their potential use for acute and chronic kidney diseases.Citation84 The discrepancy concerning renal differentiation potential of AFS cells in in vitro vs. in vivo experiments as well as AFS cell mechanism of action for kidney repair need further investigation.

Blood: hematopoiesis and hematologic diseases

Hematopoietic stem cells (HSC) lie at the top of the hematopoietic ontogeny and, if engrafted in the right niche, can theoretically reconstitute the entire blood supply. Thus, the generation of autologous HSCs from patient-specificic stem cells offers real promise for cell-therapy of genetic and malignant blood disorders. The hematopoietic potential of AFS cells was explored by Ditadi et al. in 2009.Citation37 Murine AFS cells were obtained from E12.5 to E13.5 GFP+ mice. Human AFS cells were derived from 2nd trimester amniotic fluid. CD117+/hematopoietic lineage negative cells were immediately selected after collection of the amniotic fluid. In vitro, AFS cells exhibited strong multilineage hematopoietic potential; in semisolid media, these cells were able to generate erythroid, myeloid and lymphoid colonies. Moreover, murine cells exhibit the same clonogenic potential (0.03%) as hematopoietic progenitors present in the liver at the same stage of development. In vivo, mouse AFS cells (2 × 104 cells/animal i.v. injected) were able to generate all three hematopoietic lineages after primary and secondary transplantation into immunocompromised hosts (i.e., sublethally irradiated Rag−/− mice), demonstrating their ability to self-renew. These results clearly show that AFS CD117+ cells present in the amniotic fluid have true hematopoietic potential, both in vitro and in vivo and support the idea that they may be a new source of stem cells for therapeutic applications.

Lung: AFS cell differentiation in organotypic cultures and therapeutic potential in animal models of disease

Chronic lung diseases are common and debilitating; medical therapies have restricted efficacy and lung transplantation is often the only effective treatment.Citation85 The use of stem cells for lung repair and regeneration after injury holds promise as a potential therapeutic approach for many lung diseases; however, current studies are still in their infancy.Citation86 AFS cells (human, 2nd trimester and CD117+-selected) ability to integrate into the lung and to differentiate into pulmonary lineages has been investigated in different experimental models of lung damage and development.Citation46 In vitro, AFS cells injected into mouse embryonic lung explants engrafted into the epithelium and the mesenchyme and express the early pulmonary differentiation marker TFF1. In vivo, when injected into nude mice in the absence of lung damage, systemically administered AFS cells homed to the lung but did not differentiate into specialized cells. When injected into nude mice with lung damage (i.e., hyperoxia or naphthalene injury), AFS cells not only exhibit a strong tissue engraftment but also express specific alveolar and bronchiolar epithelial markers (e.g., TFF1, SPC and CC10).Citation46 A recent work demonstrated that, in case of hyperoxia, alveolar epithelial cell type 2 (AEC2) produced chemotactic substances that increase AFS cell migration to the site of damage and that stimulate AFS cells to express phenotypic markers of distal alveolar epithelial cells (i.e., SPC, TTF-1 and ABCA3), thus leading to tissue repair.Citation87 These results illustrate AFS cell capacity of responding to lung damage by expressing specific alveolar or bronchiolar epithelial cell lineage markers, depending on the type of injury.

Nervous system: neurogenic differentiation

Stem cells represent a promising therapeutic strategy for the cure of different neurologic diseases including neurodegenerative disorders and central nervous system acute injuries.Citation88,Citation89 The demonstration that AFS cells are capable of entering the neuroectodermal lineage in neuronal conditions was given in 2007. Monoclonal CD117+ human AFS cells were induced to differentiate upon the neurogenic lineage as proven by specific marker expression (e.g., GIRK potassium channels), morphology changes, exhibition of barium-sensitive potassium current and glutamate release after stimulation.Citation35 However, CD117+ AFS cell capacity of producing mature, functional neurons in vitro and in vivo is still under investigation.Citation41,Citation90,Citation91

Discussion and Conclusions

AFS cell biological properties

The AF contains a broad range of cells, the majority of which are terminally differentiated and have limited proliferative capabilities. Since the first evidence that the AF could contain stem cells in 2003,Citation20 many groups have reported the isolation of multipotent and pluripotent cells.Citation33,Citation34 In many studies, however, mixtures of unselected, non-clonal cells have been employed,Citation90 leading to conflicting results and uncertainty regarding the identity of the cell population employed. Indeed, stem cells within the AF harbor a spectrum of potency ranging from multipotent cells to unipotent committed progenitors (e.g., CD24+OB-cadherin+ renal progenitors, SOX2+ neural progenitors and CD133+ endothelial progenitors).Citation92-Citation95 AFS cells are a subpopulation (approximately 1%) of cells within the AF expressing the type III tyrosine kinase receptor of the stem cell factor (i.e., c-kit or CD117). Since their first isolation in 2007,Citation35 succeeding studies have provided evidence concerning their broad multipotency and therapeutic potential in animal models of disease. Morphological, immunocytochemical, biochemical and molecular investigations have revealed that AFS cells represent a novel and specific entity, being distinct from other stem cell populations including those deriving from extra-embryonic tissues. AFS cells present intermediate characteristics between ES cells and mesenchymal stem cells (MSCs)Citation17,Citation96 as they, on the one hand, express embryonic markers, harbor a great proliferative and clonogenic potential, differentiate into multiple lineages and generate EBs and, on the other hand, express mesenchymal markers and do not form teratomas after implantation in vivo. Despite AFS cell capacity of differentiating into lineages representative of all embryonic germ layers and of generating EBs, their lack of tumorigenesis has been frequently advocated against their pluripotency. To this regard, the recent work of Moschidou et al.Citation38 has demonstrated that AFS cells can be reprogrammed to functional pluripotent stem cells (i.e., able to generate teratomas after injection in vivo) with the exclusive employment of a histone deacetylase inhibitor in a transgene-free approach, thus suggesting that only epigenetic modifications are required to obtain a whole pluripotent state in these cells. It is to be emphasized that this result was obtained with the exclusive employment of 1st trimester human AFS cells; further studies are therefore needed to explore eventual differences among AFS cell obtained at different gestational ages and to evaluate the efficacy of valproic acid reprogramming on 2nd trimester AFS cells. Moreover, the differentiation capacity and the tissue repair potential of AFiPS cells need to be investigated in vivo in animal models of disease.

Based on available data, AFS cells carry specific advantages in comparison to other stem cell types such as adult stem (AS) cells, ES cells, or induced pluripotent stem (iPS) cells.

(1) Obtainment

While AS cells can be difficult to isolate from their niches and ES cells derivation requires the disruption of embryos, AFS cells can be easily obtained without ethical concerns from the AF during termination procedures or diagnostic amniocenteses. Actually, the safety of amniocentesis has been established by several studies documenting an extremely low overall fetal loss rate (0.06–0.83%) related to this procedureCitation97,Citation98 and AFS cells can be obtained from AF samples without interfering with diagnostic procedures.Citation99-Citation101

(2) Safety

While ES and iPS cells are tumorigenic, AS cells can harbor genetic alterations occurring with aging and iPS cells accumulate genetic alterations during propagation in culture, AFS cells are genomically stable and do not form tumors when injected in vivo.Citation99-Citation102

(3) Reprogramming

The reprogramming of adult somatic cells is a complex process that requires the introduction of exogenous factors. First trimester AFS cells, otherwise, can be reprogrammed with high efficiency and with a stable maintenance of the pluripotent phenotype only with the employment of a chemical substance (i.e., valproic acid, an FDA-approved drug for the treatment of epilepsy), thus reducing the risks associated with the random integration of the reprogramming transgenes into the host genome.Citation103-Citation108 Many questions concerning AFS cell biological properties remain to be solved including origin, biological function in vivo, epigenetic state, and immunological reactivity. Answers to all of these issues are necessary before AFS cell employment in therapy.

AFS cell therapeutic potential

Despite the very recent identification of AFS cells, many studies have investigated their actual differentiation and therapeutic potential in vivo and in experimental models of organ development (i.e., organotypic cultures).Citation42,Citation46 Second trimester human AFS cells or rodent cells of equivalent gestational age have been employed in all cases; to our knowledge, no data resulting from the administration of 1st trimester AFS cells or reprogrammed AFiPS cells are currently available. In the majority of the existing studies AFS cells have been utilized after previous expansion in culture; only in the works by Ditadi et al.Citation37 and Piccoli et al.Citation44 the cells were administered in vivo immediately after isolation from the AF without any passage in vitro.

Pre-expanded in culture AFS cells

In organotypical cultures of embryonic organs (kidney and lung), AFS cells integrate in the developing tissue, participate to organogenesis and express markers of differentiation toward committed cells.Citation42,Citation46 In the absence of organ damage, AFS cells diffuse systemically and home to different organs but do not show signs of commitment.Citation46,Citation109 In animal models of disease, systemically administered AFS cells integrate into the damaged tissue and differentiate toward endoderm- and mesoderm-derived specialized cells (data concerning AFS cell capacity of differentiating toward the ectodermal lineage in vivo are still lacking). Despite a low efficiency of tissue engraftment, AFS cells significantly improve animal survival and reduce tissue damage both at functional and morphological levels. The common hypothesis is that AFS cells act by modulating inflammation, apoptosis as well as cell proliferation via the production of paracrine/endocrine effectors.Citation39,Citation51,Citation57,Citation61

Non-cultured AFS cells

In the work of Ditadi et al.Citation37 and Piccoli et al.Citation44 low numbers of murine AFS cells were injected in mice (around 2 × 104 cells/animal) respectively affected by severe immunodeficiency and muscle dystrophy. Freshly isolated AFS cells not only improved animal morbidity and mortality but also showed high rates of tissue engraftment and cell differentiation, thus regenerating all hematopoietic lineages and muscle fibers after primary and secondary transplantation.Citation37,Citation44 The discrepancy among results obtained with cells pre-expanded in culture vs. freshly isolated AFS cells in terms of regeneration potential may be determined by cell changes induced by culture conditions (e.g., reduced survival of the stem cell fraction, beginning of differentiation processes and modifications of cell epigenetic state). Thus, optimization of AFS cell culture conditions is mandatory in order to progress toward the therapeutic application of these cells. Although further studies are needed to better understand their biological properties and to define their therapeutic potential, AFS cells appear to be promising candidates for cell therapy and tissue engineering. In a future clinical scenario, AFS cells collected during routinely performed amniocenteses and termination procedures could be banked and, in case of need, subsequently expanded in culture or engineered in acellular grafts.Citation17,Citation110 The autologous employment of AFS cells looks particularly appealing for the treatment of perinatal disorders such as congenital malformations (e.g., congenital diaphragmatic hernia) and acquired neonatal diseases requiring tissue repair/regeneration (e.g., necrotizing enterocolitis). In this way, affected children could benefit from having autologous expanded/engineered cells ready for implantation either before birth or in the neonatal period. Moreover, considering that a bank of 150 donor cell lines would provide a beneficial match for up to 37.9% of the population,Citation111 AFS cell banks would also benefit their heterologous employment as multi/pluripotent cells of fetal origin.

Acknowledgments

M.C. is supported by Citta’ della Speranza, Malo Italy. P.D.C. is supported by the Great Ormond Street Hospital Children’s Charity.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Underwood MA, Gilbert WM, Sherman MP. Amniotic fluid: not just fetal urine anymore. J Perinatol 2005; 25:341 - 8; http://dx.doi.org/10.1038/sj.jp.7211290; PMID: 15861199
  • Klemmt PA, Vafaizadeh V, Groner B. The potential of amniotic fluid stem cells for cellular therapy and tissue engineering. Expert Opin Biol Ther 2011; 11:1297 - 314; http://dx.doi.org/10.1517/14712598.2011.587800; PMID: 21623704
  • Miki T, Strom SC. Amnion-derived pluripotent/multipotent stem cells. Stem Cell Rev 2006; 2:133 - 42; http://dx.doi.org/10.1007/s12015-006-0020-0; PMID: 17237552
  • Mescher EJ, Platzker AC, Ballard PL, Kitterman JA, Clements JA, Tooley WH. Ontogeny of tracheal fluid, pulmonary surfactant, and plasma corticoids in the fetal lamb. J Appl Physiol 1975; 39:1017 - 21; PMID: 2573
  • Le Moyec L, Muller F, Eugene M, Spraul M. Proton magnetic resonance spectroscopy of human amniotic fluids sampled at 17-18 weeks of pregnancy in cases of decreased digestive enzyme activities and detected cystic fibrosis. Clin Biochem 1994; 27:475 - 83; http://dx.doi.org/10.1016/0009-9120(94)00051-V; PMID: 7697893
  • Fauza D. Amniotic fluid and placental stem cells. Best Pract Res Clin Obstet Gynaecol 2004; 18:877 - 91; http://dx.doi.org/10.1016/j.bpobgyn.2004.07.001; PMID: 15582544
  • Dobreva MP, Pereira PN, Deprest J, Zwijsen A. On the origin of amniotic stem cells: of mice and men. Int J Dev Biol 2010; 54:761 - 77; http://dx.doi.org/10.1387/ijdb.092935md; PMID: 20446274
  • Lotgering FK, Wallenburg HC. Mechanisms of production and clearance of amniotic fluid. Semin Perinatol 1986; 10:94 - 102; PMID: 3541226
  • Gosden CM. Amniotic fluid cell types and culture. Br Med Bull 1983; 39:348 - 54; PMID: 6357346
  • Cremer M, Schachner M, Cremer T, Schmidt W, Voigtländer T. Demonstration of astrocytes in cultured amniotic fluid cells of three cases with neural-tube defect. Hum Genet 1981; 56:365 - 70; http://dx.doi.org/10.1007/BF00274694; PMID: 7016720
  • Cremer M, Treiss I, Cremer T, Hager D, Franke WW. Characterization of cells of amniotic fluids by immunological identification of intermediate-sized filaments: presence of cells of different tissue origin. Hum Genet 1981; 59:373 - 9; http://dx.doi.org/10.1007/BF00295475; PMID: 6174407
  • Johnston P, Salk D, Martin GM, Hoehn H. Cultivated cells from mid-trimester amniotic fluids: IV. Cell type indentification via one and two-dimensional electrophoresis of clonal whole cell homogenates. Prenat Diagn 1982; 2:79 - 88; http://dx.doi.org/10.1002/pd.1970020202; PMID: 7145843
  • Milunsky A, Blusztajn JK, Zeisel SH. Amniotic-fluid total cholinesterase and neural-tube defects. Lancet 1979; 2:36; http://dx.doi.org/10.1016/S0140-6736(79)90196-X; PMID: 87905
  • Cananzi M, Atala A, De Coppi P. Stem cells derived from amniotic fluid: new potentials in regenerative medicine. Reprod Biomed Online 2009; 18:Suppl 1 17 - 27; http://dx.doi.org/10.1016/S1472-6483(10)60111-3; PMID: 19281660
  • Hoehn H, Bryant EM, Fantel AG, Martin GM. Cultivated cells from diagnostic amniocentesis in second trimester pregnancies. III. The fetal urine as a potential source of clonable cells. Humangenetik 1975; 29:285 - 90; http://dx.doi.org/10.1007/BF00394190; PMID: 1176144
  • Gosden C, Brock DJ. Amniotic fluid cell morphology in early antenatal prediction of abortion and low birth weight. Br Med J 1978; 2:1186 - 9; http://dx.doi.org/10.1136/bmj.2.6146.1186; PMID: 82463
  • Siegel N, Rosner M, Hanneder M, Valli A, Hengstschläger M. Stem cells in amniotic fluid as new tools to study human genetic diseases. Stem Cell Rev 2007; 3:256 - 64; http://dx.doi.org/10.1007/s12015-007-9003-z; PMID: 17955390
  • Torricelli F, Brizzi L, Bernabei PA, Gheri G, Di Lollo S, Nutini L, et al. Identification of hematopoietic progenitor cells in human amniotic fluid before the 12th week of gestation. Ital J Anat Embryol 1993; 98:119 - 26; PMID: 8239855
  • Streubel B, Martucci-Ivessa G, Fleck T, Bittner RE. In vitro transformation of amniotic cells to muscle cells–background and outlook. Wien Med Wochenschr 1996; 146:216 - 7; PMID: 9012220
  • Prusa AR, Marton E, Rosner M, Bernaschek G, Hengstschläger M. Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research?. Hum Reprod 2003; 18:1489 - 93; http://dx.doi.org/10.1093/humrep/deg279; PMID: 12832377
  • Schöler HR, Hatzopoulos AK, Balling R, Suzuki N, Gruss P. A family of octamer-specific proteins present during mouse embryogenesis: evidence for germline-specific expression of an Oct factor. EMBO J 1989; 8:2543 - 50; PMID: 2573523
  • Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet 2000; 24:372 - 6; http://dx.doi.org/10.1038/74199; PMID: 10742100
  • Donovan PJ, De Miguel MP, Hirano MP, Parsons MS, Lincoln AJ. Germ cell biology–from generation to generation. Int J Dev Biol 2001; 45:523 - 31; PMID: 11417895
  • Pesce M, Schöler HR. Oct-4: gatekeeper in the beginnings of mammalian development. Stem Cells 2001; 19:271 - 8; http://dx.doi.org/10.1634/stemcells.19-4-271; PMID: 11463946
  • Gidekel S, Pizov G, Bergman Y, Pikarsky E. Oct-3/4 is a dose-dependent oncogenic fate determinant. Cancer Cell 2003; 4:361 - 70; http://dx.doi.org/10.1016/S1535-6108(03)00270-8; PMID: 14667503
  • Looijenga LH, Stoop H, de Leeuw HP, de Gouveia Brazao CA, Gillis AJ, van Roozendaal KE, et al. POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors. Cancer Res 2003; 63:2244 - 50; PMID: 12727846
  • Berg JS, Goodell MA. An argument against a role for Oct4 in somatic stem cells. Cell Stem Cell 2007; 1:359 - 60; http://dx.doi.org/10.1016/j.stem.2007.09.007; PMID: 18371372
  • Lengner CJ, Camargo FD, Hochedlinger K, Welstead GG, Zaidi S, Gokhale S, et al. Oct4 expression is not required for mouse somatic stem cell self-renewal. Cell Stem Cell 2007; 1:403 - 15; http://dx.doi.org/10.1016/j.stem.2007.07.020; PMID: 18159219
  • Liedtke S, Enczmann J, Waclawczyk S, Wernet P, Kögler G. Oct4 and its pseudogenes confuse stem cell research. Cell Stem Cell 2007; 1:364 - 6; http://dx.doi.org/10.1016/j.stem.2007.09.003; PMID: 18371374
  • Bossolasco P, Montemurro T, Cova L, Zangrossi S, Calzarossa C, Buiatiotis S, et al. Molecular and phenotypic characterization of human amniotic fluid cells and their differentiation potential. Cell Res 2006; 16:329 - 36; http://dx.doi.org/10.1038/sj.cr.7310043; PMID: 16617328
  • Stefanidis K, Loutradis D, Anastasiadou V, Bletsa R, Kiapekou E, Drakakis P, et al. Oxytocin receptor- and Oct-4-expressing cells in human amniotic fluid. Gynecol Endocrinol 2008; 24:280 - 4; http://dx.doi.org/10.1080/09513590801977167; PMID: 18569033
  • Karlmark KR, Freilinger A, Marton E, Rosner M, Lubec G, Hengstschläger M. Activation of ectopic Oct-4 and Rex-1 promoters in human amniotic fluid cells. Int J Mol Med 2005; 16:987 - 92; PMID: 16273276
  • Tsai MS, Hwang SM, Tsai YL, Cheng FC, Lee JL, Chang YJ. Clonal amniotic fluid-derived stem cells express characteristics of both mesenchymal and neural stem cells. Biol Reprod 2006; 74:545 - 51; http://dx.doi.org/10.1095/biolreprod.105.046029; PMID: 16306422
  • Kim J, Lee Y, Kim H, Hwang KJ, Kwon HC, Kim SK, et al. Human amniotic fluid-derived stem cells have characteristics of multipotent stem cells. Cell Prolif 2007; 40:75 - 90; http://dx.doi.org/10.1111/j.1365-2184.2007.00414.x; PMID: 17227297
  • De Coppi P, Bartsch G Jr., Siddiqui MM, Xu T, Santos CC, Perin L, et al. Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 2007; 25:100 - 6; http://dx.doi.org/10.1038/nbt1274; PMID: 17206138
  • Zsebo KM, Williams DA, Geissler EN, Broudy VC, Martin FH, Atkins HL, et al. Stem cell factor is encoded at the Sl locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor. Cell 1990; 63:213 - 24; http://dx.doi.org/10.1016/0092-8674(90)90302-U; PMID: 1698556
  • Ditadi A, de Coppi P, Picone O, Gautreau L, Smati R, Six E, et al. Human and murine amniotic fluid c-Kit+Lin- cells display hematopoietic activity. Blood 2009; 113:3953 - 60; http://dx.doi.org/10.1182/blood-2008-10-182105; PMID: 19221036
  • Moschidou D, Mukherjee S, Blundell MP, Drews K, Jones GN, Abdulrazzak H, et al. Valproic Acid Confers Functional Pluripotency to Human Amniotic Fluid Stem Cells in a Transgene-free Approach. Mol Ther 2012; 20:1953 - 67; http://dx.doi.org/10.1038/mt.2012.117; PMID: 22760542
  • Sedrakyan S, Da Sacco S, Milanesi A, Shiri L, Petrosyan A, Varimezova R, et al. Injection of amniotic fluid stem cells delays progression of renal fibrosis. J Am Soc Nephrol 2012; 23:661 - 73; http://dx.doi.org/10.1681/ASN.2011030243; PMID: 22302195
  • Bollini S, Pozzobon M, Nobles M, Riegler J, Dong X, Piccoli M, et al. In vitro and in vivo cardiomyogenic differentiation of amniotic fluid stem cells. Stem Cell Rev 2011; 7:364 - 80; http://dx.doi.org/10.1007/s12015-010-9200-z; PMID: 21120638
  • Arnhold S, Glüer S, Hartmann K, Raabe O, Addicks K, Wenisch S, et al. Amniotic-Fluid Stem Cells: Growth Dynamics and Differentiation Potential after a CD-117-Based Selection Procedure. Stem Cells Int 2011; 2011:715341; http://dx.doi.org/10.4061/2011/715341; PMID: 21437196
  • Perin L, Giuliani S, Jin D, Sedrakyan S, Carraro G, Habibian R, et al. Renal differentiation of amniotic fluid stem cells. Cell Prolif 2007; 40:936 - 48; http://dx.doi.org/10.1111/j.1365-2184.2007.00478.x; PMID: 18021180
  • Valli A, Rosner M, Fuchs C, Siegel N, Bishop CE, Dolznig H, et al. Embryoid body formation of human amniotic fluid stem cells depends on mTOR. Oncogene 2010; 29:966 - 77; http://dx.doi.org/10.1038/onc.2009.405; PMID: 19935716
  • Piccoli M, Franzin C, Bertin E, Urbani L, Blaauw B, Repele A, et al. Amniotic fluid stem cells restore the muscle cell niche in a HSA-Cre, Smn(F7/F7) mouse model. Stem Cells 2012; 30:1675 - 84; http://dx.doi.org/10.1002/stem.1134; PMID: 22644669
  • Siegel N, Rosner M, Unbekandt M, Fuchs C, Slabina N, Dolznig H, et al. Contribution of human amniotic fluid stem cells to renal tissue formation depends on mTOR. Hum Mol Genet 2010; 19:3320 - 31; http://dx.doi.org/10.1093/hmg/ddq236; PMID: 20542987
  • Carraro G, Perin L, Sedrakyan S, Giuliani S, Tiozzo C, Lee J, et al. Human amniotic fluid stem cells can integrate and differentiate into epithelial lung lineages. Stem Cells 2008; 26:2902 - 11; http://dx.doi.org/10.1634/stemcells.2008-0090; PMID: 18719226
  • Chiavegato A, Bollini S, Pozzobon M, Callegari A, Gasparotto L, Taiani J, et al. Human amniotic fluid-derived stem cells are rejected after transplantation in the myocardium of normal, ischemic, immuno-suppressed or immuno-deficient rat. J Mol Cell Cardiol 2007; 42:746 - 59; http://dx.doi.org/10.1016/j.yjmcc.2006.12.008; PMID: 17300799
  • Gekas J, Walther G, Skuk D, Bujold E, Harvey I, Bertrand OF. In vitro and in vivo study of human amniotic fluid-derived stem cell differentiation into myogenic lineage. Clin Exp Med 2010; 10:1 - 6; http://dx.doi.org/10.1007/s10238-009-0060-2; PMID: 19730985
  • Ma X, Zhang S, Zhou J, Chen B, Shang Y, Gao T, et al. Clone-derived human AF-amniotic fluid stem cells are capable of skeletal myogenic differentiation in vitro and in vivo. J Tissue Eng Regen Med 2012; 6:598 - 613; http://dx.doi.org/10.1002/term.462; PMID: 22396316
  • Guan X, Delo DM, Atala A, Soker S. In vitro cardiomyogenic potential of human amniotic fluid stem cells. J Tissue Eng Regen Med 2011; 5:220 - 8; http://dx.doi.org/10.1002/term.308; PMID: 20687122
  • Rota C, Imberti B, Pozzobon M, Piccoli M, De Coppi P, Atala A, et al. Human amniotic fluid stem cell preconditioning improves their regenerative potential. Stem Cells Dev 2012; 21:1911 - 23; http://dx.doi.org/10.1089/scd.2011.0333; PMID: 22066606
  • Chen WQ, Siegel N, Li L, Pollak A, Hengstschläger M, Lubec G. Variations of protein levels in human amniotic fluid stem cells CD117/2 over passages 5-25. J Proteome Res 2009; 8:5285 - 95; http://dx.doi.org/10.1021/pr900630s; PMID: 19791749
  • Solter D. From teratocarcinomas to embryonic stem cells and beyond: a history of embryonic stem cell research. Nat Rev Genet 2006; 7:319 - 27; http://dx.doi.org/10.1038/nrg1827; PMID: 16534514
  • Itskovitz-Eldor J, Schuldiner M, Karsenti D, Eden A, Yanuka O, Amit M, et al. Differentiation of human embryonic stem cells into embryoid bodies compromising the three embryonic germ layers. Mol Med 2000; 6:88 - 95; PMID: 10859025
  • Fuchs C, Rosner M, Dolznig H, Mikula M, Kramer N, Hengstschläger M. Tuberin and PRAS40 are anti-apoptotic gatekeepers during early human amniotic fluid stem-cell differentiation. Hum Mol Genet 2012; 21:1049 - 61; http://dx.doi.org/10.1093/hmg/ddr535; PMID: 22090422
  • Siegel N, Valli A, Fuchs C, Rosner M, Hengstschläger M. Expression of mTOR pathway proteins in human amniotic fluid stem cells. Int J Mol Med 2009; 23:779 - 84; PMID: 19424604
  • Perin L, Sedrakyan S, Giuliani S, Da Sacco S, Carraro G, Shiri L, et al. Protective effect of human amniotic fluid stem cells in an immunodeficient mouse model of acute tubular necrosis. PLoS One 2010; 5:e9357; http://dx.doi.org/10.1371/journal.pone.0009357; PMID: 20195358
  • Yao M, Dieterle T, Hale SL, Dow JS, Kedes LH, Peterson KL, et al. Long-term outcome of fetal cell transplantation on postinfarction ventricular remodeling and function. J Mol Cell Cardiol 2003; 35:661 - 70; http://dx.doi.org/10.1016/S0022-2828(03)00098-1; PMID: 12788384
  • Dai W, Kloner RA. Myocardial regeneration by human amniotic fluid stem cells: challenges to be overcome. J Mol Cell Cardiol 2007; 42:730 - 2; http://dx.doi.org/10.1016/j.yjmcc.2007.01.013; PMID: 17362984
  • Delo DM, Guan X, Wang Z, Groban L, Callahan M, Smith T, et al. Calcification after myocardial infarction is independent of amniotic fluid stem cell injection. Cardiovasc Pathol 2011; 20:e69 - 78; http://dx.doi.org/10.1016/j.carpath.2010.03.001; PMID: 20382039
  • Bollini S, Cheung KK, Riegler J, Dong X, Smart N, Ghionzoli M, et al. Amniotic fluid stem cells are cardioprotective following acute myocardial infarction. Stem Cells Dev 2011; 20:1985 - 94; http://dx.doi.org/10.1089/scd.2010.0424; PMID: 21534857
  • Price FD, Kuroda K, Rudnicki MA. Stem cell based therapies to treat muscular dystrophy. Biochim Biophys Acta 2007; 1772:272 - 83; http://dx.doi.org/10.1016/j.bbadis.2006.08.011; PMID: 17034994
  • Meregalli M, Farini A, Colleoni F, Cassinelli L, Torrente Y. The role of stem cells in muscular dystrophies. Curr Gene Ther 2012; 12:192 - 205; http://dx.doi.org/10.2174/156652312800840559; PMID: 22463740
  • Beardi J, Hessmann M, Hansen M, Rommens PM. Operative treatment of tibial shaft fractures: a comparison of different methods of primary stabilisation. Arch Orthop Trauma Surg 2008; 128:709 - 15; http://dx.doi.org/10.1007/s00402-008-0619-5; PMID: 18465138
  • Salgado AJ, Oliveira JT, Pedro AJ, Reis RL. Adult stem cells in bone and cartilage tissue engineering. Curr Stem Cell Res Ther 2006; 1:345 - 64; PMID: 18220879
  • Bianco P, Robey PG. Stem cells in tissue engineering. Nature 2001; 414:118 - 21; http://dx.doi.org/10.1038/35102181; PMID: 11689957
  • Peister A, Deutsch ER, Kolambkar Y, Hutmacher DW, Guldberg RE. Amniotic fluid stem cells produce robust mineral deposits on biodegradable scaffolds. Tissue Eng Part A 2009; 15:3129 - 38; http://dx.doi.org/10.1089/ten.tea.2008.0536; PMID: 19344289
  • Maraldi T, Riccio M, Resca E, Pisciotta A, La Sala GB, Ferrari A, et al. Human amniotic fluid stem cells seeded in fibroin scaffold produce in vivo mineralized matrix. Tissue Eng Part A 2011; 17:2833 - 43; http://dx.doi.org/10.1089/ten.tea.2011.0062; PMID: 21864161
  • Peister A, Woodruff MA, Prince JJ, Gray DP, Hutmacher DW, Guldberg RE. Cell sourcing for bone tissue engineering: amniotic fluid stem cells have a delayed, robust differentiation compared to mesenchymal stem cells. Stem Cell Res 2011; 7:17 - 27; http://dx.doi.org/10.1016/j.scr.2011.03.001; PMID: 21531647
  • Sun H, Feng K, Hu J, Soker S, Atala A, Ma PX. Osteogenic differentiation of human amniotic fluid-derived stem cells induced by bone morphogenetic protein-7 and enhanced by nanofibrous scaffolds. Biomaterials 2010; 31:1133 - 9; http://dx.doi.org/10.1016/j.biomaterials.2009.10.030; PMID: 19857889
  • Rodrigues MT, Lee BK, Lee SJ, Gomes ME, Reis RL, Atala A, et al. The effect of differentiation stage of amniotic fluid stem cells on bone regeneration. Biomaterials 2012; 33:6069 - 78; http://dx.doi.org/10.1016/j.biomaterials.2012.05.016; PMID: 22672834
  • Riccio M, Maraldi T, Pisciotta A, La Sala GB, Ferrari A, Bruzzesi G, et al. Fibroin scaffold repairs critical-size bone defects in vivo supported by human amniotic fluid and dental pulp stem cells. Tissue Eng Part A 2012; 18:1006 - 13; http://dx.doi.org/10.1089/ten.tea.2011.0542; PMID: 22166080
  • Koelling S, Miosge N. Stem cell therapy for cartilage regeneration in osteoarthritis. Expert Opin Biol Ther 2009; 9:1399 - 405; http://dx.doi.org/10.1517/14712590903246370; PMID: 19793003
  • Kolambkar YM, Peister A, Soker S, Atala A, Guldberg RE. Chondrogenic differentiation of amniotic fluid-derived stem cells. J Mol Histol 2007; 38:405 - 13; http://dx.doi.org/10.1007/s10735-007-9118-1; PMID: 17668282
  • Bussolati B, Hauser PV, Carvalhosa R, Camussi G. Contribution of stem cells to kidney repair. Curr Stem Cell Res Ther 2009; 4:2 - 8; http://dx.doi.org/10.2174/157488809787169129; PMID: 19149624
  • Bruce SJ, Rea RW, Steptoe AL, Busslinger M, Bertram JF, Perkins AC. In vitro differentiation of murine embryonic stem cells toward a renal lineage. Differentiation 2007; 75:337 - 49; http://dx.doi.org/10.1111/j.1432-0436.2006.00149.x; PMID: 17286599
  • Bruno S, Bussolati B, Grange C, Collino F, di Cantogno LV, Herrera MB, et al. Isolation and characterization of resident mesenchymal stem cells in human glomeruli. Stem Cells Dev 2009; 18:867 - 80; http://dx.doi.org/10.1089/scd.2008.0320; PMID: 19579288
  • Fuente Mora C, Ranghini E, Bruno S, Bussolati B, Camussi G, Wilm B, et al. Differentiation of podocyte and proximal tubule-like cells from a mouse kidney-derived stem cell line. Stem Cells Dev 2012; 21:296 - 307; http://dx.doi.org/10.1089/scd.2010.0470; PMID: 21510739
  • Kramer J, Steinhoff J, Klinger M, Fricke L, Rohwedel J. Cells differentiated from mouse embryonic stem cells via embryoid bodies express renal marker molecules. Differentiation 2006; 74:91 - 104; http://dx.doi.org/10.1111/j.1432-0436.2006.00062.x; PMID: 16533308
  • Morigi M, Introna M, Imberti B, Corna D, Abbate M, Rota C, et al. Human bone marrow mesenchymal stem cells accelerate recovery of acute renal injury and prolong survival in mice. Stem Cells 2008; 26:2075 - 82; http://dx.doi.org/10.1634/stemcells.2007-0795; PMID: 18499895
  • Morigi M, Rota C, Montemurro T, Montelatici E, Lo Cicero V, Imberti B, et al. Life-sparing effect of human cord blood-mesenchymal stem cells in experimental acute kidney injury. Stem Cells 2010; 28:513 - 22; PMID: 20049901
  • Murray P, Camussi G, Davies JA, Edgar D, Hengstschlager M, Kenny S, et al. The KIDSTEM European Research Training Network: Developing a Stem Cell Based Therapy to Replace Nephrons Lost through Reflux Nephropathy. Organogenesis 2007; 3:2 - 5; http://dx.doi.org/10.4161/org.3.1.3440; PMID: 19279693
  • Hauser PV, De Fazio R, Bruno S, Sdei S, Grange C, Bussolati B, et al. Stem cells derived from human amniotic fluid contribute to acute kidney injury recovery. Am J Pathol 2010; 177:2011 - 21; http://dx.doi.org/10.2353/ajpath.2010.091245; PMID: 20724594
  • Rosner M, Schipany K, Gundacker C, Shanmugasundaram B, Li K, Fuchs C, et al. Renal differentiation of amniotic fluid stem cells: perspectives for clinical application and for studies on specific human genetic diseases. Eur J Clin Invest 2012; 42:677 - 84; http://dx.doi.org/10.1111/j.1365-2362.2011.02622.x; PMID: 22060053
  • Loebinger MR, Aguilar S, Janes SM. Therapeutic potential of stem cells in lung disease: progress and pitfalls. Clin Sci (Lond) 2008; 114:99 - 108; http://dx.doi.org/10.1042/CS20070073; PMID: 18062775
  • Moodley Y, Manuelpillai U, Weiss DJ. Cellular therapies for lung disease: a distant horizon. Respirology 2011; 16:223 - 37; http://dx.doi.org/10.1111/j.1440-1843.2010.01914.x; PMID: 21199158
  • Buckley S, Shi W, Carraro G, Sedrakyan S, Da Sacco S, Driscoll BA, et al. The milieu of damaged alveolar epithelial type 2 cells stimulates alveolar wound repair by endogenous and exogenous progenitors. Am J Respir Cell Mol Biol 2011; 45:1212 - 21; http://dx.doi.org/10.1165/rcmb.2010-0325OC; PMID: 21700959
  • Carney BJ, Shah K. Migration and fate of therapeutic stem cells in different brain disease models. Neuroscience 2011; 197:37 - 47; http://dx.doi.org/10.1016/j.neuroscience.2011.08.063; PMID: 21946010
  • Decimo I, Bifari F, Krampera M, Fumagalli G. Neural stem cell niches in health and diseases. Curr Pharm Des 2012; 18:1755 - 83; http://dx.doi.org/10.2174/138161212799859611; PMID: 22394166
  • Rosner M, Schipany K, Shanmugasundaram B, Lubec G, Hengstschläger M. Amniotic fluid stem cells: future perspectives. Stem Cells Int 2012; 2012:741810; http://dx.doi.org/10.1155/2012/741810; PMID: 22719774
  • Toselli M, Cerbai E, Rossi F, Cattaneo E. Do amniotic fluid-derived stem cells differentiate into neurons in vitro?. Nat Biotechnol 2008; 26:269 - 70, author reply 270-1; http://dx.doi.org/10.1038/nbt0308-269; PMID: 18327228
  • 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 2010; In press PMID: 21157345
  • Da Sacco S, Sedrakyan S, Boldrin F, Giuliani S, Parnigotto P, Habibian R, et al. Human amniotic fluid as a potential new source of organ specific precursor cells for future regenerative medicine applications. J Urol 2010; 183:1193 - 200; http://dx.doi.org/10.1016/j.juro.2009.11.006; PMID: 20096867
  • Jiang TM, Yang ZJ, Kong CZ, Zhang HT. Schwann-like cells can be induction from human nestin-positive amniotic fluid mesenchymal stem cells. In Vitro Cell Dev Biol Anim 2010; 46:793 - 800; http://dx.doi.org/10.1007/s11626-010-9335-x; PMID: 20652439
  • Schmidt D, Achermann J, Odermatt B, Breymann C, Mol A, Genoni M, et al. Prenatally fabricated autologous human living heart valves based on amniotic fluid derived progenitor cells as single cell source. Circulation 2007; 116:Suppl I64 - 70; http://dx.doi.org/10.1161/CIRCULATIONAHA.106.681494; PMID: 17846327
  • Bajada S, Mazakova I, Richardson JB, Ashammakhi N. Updates on stem cells and their applications in regenerative medicine. J Tissue Eng Regen Med 2008; 2:169 - 83; http://dx.doi.org/10.1002/term.83; PMID: 18493906
  • Caughey AB, Hopkins LM, Norton ME. Chorionic villus sampling compared with amniocentesis and the difference in the rate of pregnancy loss. Obstet Gynecol 2006; 108:612 - 6; http://dx.doi.org/10.1097/01.AOG.0000232512.46869.fc; PMID: 16946222
  • Eddleman KA, Malone FD, Sullivan L, Dukes K, Berkowitz RL, Kharbutli Y, et al. Pregnancy loss rates after midtrimester amniocentesis. Obstet Gynecol 2006; 108:1067 - 72; http://dx.doi.org/10.1097/01.AOG.0000240135.13594.07; PMID: 17077226
  • Daley GQ, Ahrlund Richter L, Auerbach JM, Benvenisty N, Charo RA, Chen G, et al. Ethics. The ISSCR guidelines for human embryonic stem cell research. Science 2007; 315:603 - 4; http://dx.doi.org/10.1126/science.1139337; PMID: 17272706
  • Edwards RG. A burgeoning science of embryological genetics demands a modern ethics. Reprod Biomed Online 2007; 15:Suppl 1 34 - 40; http://dx.doi.org/10.1016/S1472-6483(10)60356-2; PMID: 17822615
  • Mimeault M, Hauke R, Mehta PP, Batra SK. Recent advances in cancer stem/progenitor cell research: therapeutic implications for overcoming resistance to the most aggressive cancers. J Cell Mol Med 2007; 11:981 - 1011; http://dx.doi.org/10.1111/j.1582-4934.2007.00088.x; PMID: 17979879
  • Laurent LC, Ulitsky I, Slavin I, Tran H, Schork A, Morey R, et al. Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPSCs during reprogramming and time in culture. Cell Stem Cell 2011; 8:106 - 18; http://dx.doi.org/10.1016/j.stem.2010.12.003; PMID: 21211785
  • Kaji K, Norrby K, Paca A, Mileikovsky M, Mohseni P, Woltjen K. Virus-free induction of pluripotency and subsequent excision of reprogramming factors. Nature 2009; 458:771 - 5; http://dx.doi.org/10.1038/nature07864; PMID: 19252477
  • Kim D, Kim CH, Moon JI, Chung YG, Chang MY, Han BS, et al. Generation of human induced pluripotent stem cells by direct delivery of reprogramming proteins. Cell Stem Cell 2009; 4:472 - 6; http://dx.doi.org/10.1016/j.stem.2009.05.005; PMID: 19481515
  • Mukherjee S, Thrasher AJ. iPSCs: Unstable origins?. Mol Ther 2011; 19:1188 - 90; http://dx.doi.org/10.1038/mt.2011.116; PMID: 21720379
  • Narsinh KH, Jia F, Robbins RC, Kay MA, Longaker MT, Wu JC. Generation of adult human induced pluripotent stem cells using nonviral minicircle DNA vectors. Nat Protoc 2011; 6:78 - 88; http://dx.doi.org/10.1038/nprot.2010.173; PMID: 21212777
  • Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006; 126:663 - 76; http://dx.doi.org/10.1016/j.cell.2006.07.024; PMID: 16904174
  • Warren L, Manos PD, Ahfeldt T, Loh YH, Li H, Lau F, et al. Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA. Cell Stem Cell 2010; 7:618 - 30; http://dx.doi.org/10.1016/j.stem.2010.08.012; PMID: 20888316
  • Ghionzoli M, Cananzi M, Zani A, Rossi CA, Leon FF, Pierro A, et al. Amniotic fluid stem cell migration after intraperitoneal injection in pup rats: implication for therapy. Pediatr Surg Int 2010; 26:79 - 84; http://dx.doi.org/10.1007/s00383-009-2504-x; PMID: 19855985
  • Kunisaki SM, Armant M, Kao GS, Stevenson K, Kim H, Fauza DO. Tissue engineering from human mesenchymal amniocytes: a prelude to clinical trials. J Pediatr Surg 2007; 42:974 - 9, discussion 979-80; http://dx.doi.org/10.1016/j.jpedsurg.2007.01.031; PMID: 17560205
  • Taylor CJ, Bolton EM, Pocock S, Sharples LD, Pedersen RA, Bradley JA. Banking on human embryonic stem cells: estimating the number of donor cell lines needed for HLA matching. Lancet 2005; 366:2019 - 25; http://dx.doi.org/10.1016/S0140-6736(05)67813-0; PMID: 16338451

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.