Placenta as a Source of Stem Cells for Regenerative Medicine ...

2 downloads 0 Views 201KB Size Report
Feb 3, 2015 - Placenta derived stem cellsMesenchymal stem cellsHematopoietic stem cellsCell-based therapyRegenerative medicine. This article is part of ...
Curr Pathobiol Rep (2015) 3:9–16 DOI 10.1007/s40139-015-0070-6 TISSUE ENGINEERING AND REGENERATION, (BRYAN BROWN AND CHRISTOPHER DEARTH, SECTION EDITORS)

Placenta as a Source of Stem Cells for Regenerative Medicine Jason A. Meierhenry • Volodymyr Ryzhuk • Maricel G. Miguelino • Lee Lankford • Jerry S. Powell • Diana Farmer • Aijun Wang

Published online: 3 February 2015  Springer Science+Business Media New York 2015

Abstract The development of effective cell transplantation therapies is currently the focus of biomedical research worldwide, and numerous cell types and sources have been explored for applications in regenerative medicine. The placenta is a unique organ of fetomaternal origin that plays an important role in fetal development, and multiple types of placenta-derived stem cells show great promise for application in regenerative medicine. This review concisely summarizes the recent proceedings on the characterization, biological properties, and applications of mesenchymal stem cells that have been isolated from various anatomic regions of the placenta and at different gestational ages. In addition, this review summarizes recent studies that have isolated amniotic epithelial cells, hematopoietic stem cells, and trophoblast stem cells from placental tissue. Keywords Placenta derived stem cells  Mesenchymal stem cells  Hematopoietic stem cells  Cell-based therapy  Regenerative medicine

This article is part of the Topical Collection on Tissue Engineering and Regeneration. J. A. Meierhenry  V. Ryzhuk  L. Lankford  D. Farmer  A. Wang (&) Department of Surgery, Surgical Bioengineering Laboratory, University of California, Davis Medical Center, Sacramento, CA 95817, USA e-mail: [email protected] M. G. Miguelino  J. S. Powell Division of Hematology/Oncology, Department of Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817, USA

Introduction Over the past few decades, cellular transplantation therapy has emerged as a potential tool for tissue repair and regenerative medicine. Pre-clinical studies have shown promising results for the treatment of a wide range of degenerative diseases, and currently, investigators continue to push forward the application of cell therapy into clinical trials. However, in spite of these advancements, questions still remain in regard to which cell types and tissue sources are most beneficial for specific therapeutic applications (i.e., replacement of damaged tissue, modulation of endogenous body responses, etc.). Bone marrow has typically been the primary source of mesenchymal stem cells (MSCs) for both experimental and clinical studies; however, the low number of MSCs and the invasive procedure associated with obtaining bone marrow-derived MSCs (BM-MSCs) can make their use problematic [1]. More recently, gestational tissues, such as the placenta, placental membranes, umbilical cord, and amniotic fluid, have been recognized as potential cell sources for regenerative medicine [1]. Placental tissues in particular have been shown to be a rich source of stem cells with strong immunosuppressive properties, and these tissues demonstrate strong potential for cell therapy. The placenta is a unique organ of fetomaternal origin that facilitates nutrient and gas exchange between fetal and maternal blood. It is considered ‘‘extra-embryonic,’’ in that it develops outside of the developing fetus during gestation and will eventually become inoperative when the fetus is born and begins breathing on its own. As a fetomaternal organ, the placenta develops from the trophectoderm layer of the fetal blastocyst. The trophectoderm layer is crucial for the implantation of the embryo into maternal uterine tissue and is needed for decidualization of the endometrial

123

10

Curr Pathobiol Rep (2015) 3:9–16

layer of the uterus. During gestation, the placenta develops into discrete anatomic regions beginning from the decidua basalis (located most proximal to the uterus) and terminating at the umbilical cord. Each of these regions contains a variety of cell types that can potentially be used for cell therapy. MSCs can be readily isolated from essentially all of the anatomic regions of the placenta, with the amniotic membrane serving as a source for obtaining epithelial cells and the chorionic villi serving as a source for hematopoietic stem cells (HSCs) and trophoblasts. The placenta is partially comprised of fetal tissue, which allows for the development of therapies autologous to the fetus for in utero treatment of congenital anomalies. Due to their developmental significance, fetal cells may possess unique properties that other adult primary cell types lack. The means of obtaining placenta-derived cells may also be minimally invasive compared to other candidate tissue sources, such as bone marrow or adipose tissue. For example, chorionic villi from early gestation placenta can be readily obtained using chorionic villus sampling (CVS), which is a technique generally utilized to sample fetal DNA for prenatal diagnosis. In addition, term placenta is commonly discarded after birth and can be collected as a non-invasive tissue source for cells. For these reasons, utilizing the placenta as a source of cells for cellular therapy shows promise for targeted therapies for numerous pathological conditions. This review will cover the characterization, biological properties, specific tissue sources, and applications of placenta-derived mesenchymal stem cells (PMSCs), as well as recent studies on amniotic epithelial cells (AEC), HSCs, and trophoblast stem cells (TSCs) that have been isolated from placental tissue.

utilized stem cells in translational studies and are expected to be in high demand by clinical therapies in the near future. According to www.clinicaltrials.gov, a publicly available database with information on publicly and privately supported clinical studies, there are currently over 450 registered clinical trials that employ MSC-based therapies. MSCs can be successfully isolated from a number of tissues, including bone marrow, adipose tissue, muscle, lungs, tooth buds, umbilical cord blood, Wharton’s jelly, amniotic fluid, and different regions of the placenta [5, 6]. Depending on the tissue of origin, MSCs exhibit varied degrees of plasticity, rate of division, and secretion. A number of recent publications suggest that placenta- and amnion-derived mesenchymal stem cells have a wide range of cell-based therapeutic applications, including pancreatic and liver regeneration, neurological diseases, and myocardial and lung rescue cell therapies [5, 7–9]. Phenotype of PMSCs

PMSCs

Both BM-MSCs and PMSCs have been demonstrated to adhere to plastic and possess morphology similar to fibroblasts [10]. Overall, the size and shape of PMSCs are more heterogeneous than stem cells derived from bone marrow. The human PMSC surface marker phenotype is consistent with adult MSCs, indicated by the presence of CD44, CD73, CD90, and CD105 and by the absence of CD11, CD45, and CD79. Gene expression profiles are not significantly altered within differently sourced MSCs [9]; however, the expression levels of specific factors vary depending on the source of cells. For instance, the level of hepatocyte growth factor expression, a cytokine primarily responsible for angiogenesis and many other biological functions, was shown to be much higher in PMSCs when compared to adult MSCs [11].

Introduction to MSCs

Differentiation of PMSCs

In order to be classified as MSCs, cells must meet the following criteria [2]:

Both PMSCs and BM-MSCs show tri-lineage differentiation potential (the ability to differentiate into chondrocytes, adipocytes, and osteocytes), but the efficiency of differentiation varies depending on the source of stem cells [10]. Studies have found that PMSCs are less efficient than BMMSCs at differentiating into adipocytes and osteocytes [12]. On the other hand, PMSCs are ontogenically related to embryonic stem cells, sharing their immunoprivileged characteristics and surpassing adult MSCs in broader plasticity and augmented proliferation rate [13]. For example, amnion-derived mesenchymal stem cells (AMSCs) express the pluripotent genetic marker OCT-4, do not form tumors in vivo, and can be successfully induced to differentiate into six lineages (myogenic, adipogenic, osteogenic, endothelial, hepatic and neuronal) [14].

– –



Being capable of adhering to plastic in standard culture conditions. Expressing characteristic cell surface markers such as CD73, CD90, and CD105 and lacking the expression of CD11b/CD14, CD34, CD45, CD79a/CD19, and HLADR markers. Being capable of differentiating into chondroblasts, osteoblasts, and adipocytes.

MSCs were first derived from bone marrow stroma, and BM-MSCs have been considered the gold standard for MSCs due to the relative ease of isolation, extended plasticity, and proven clinical applications [3, 4]. MSCs are one of the most

123

Curr Pathobiol Rep (2015) 3:9–16

Proliferation of PMSCs Barlow et al. demonstrated higher growth rate of PMSCs than BM-MSCs. PMSCs were able to withstand higher number of passages and lower seeding density than BMMSCs in culture [10]. AMSCs are highly proliferative and clonogenic and have the potential to expand for 15 passages [15]. MSCs obtained from preterm placenta maintain long telomeres and express markers associated with pluripotency, such as OCT-4 and SSEA-3 [14]. Secretion of PMSCs Currently, studies are exploring the potential of MSCs to replace cells of damaged tissue, and investigators are interested in the ability of MSCs to modulate the local environment by releasing chemokines, cytokines, and prostaglandins that may promote vascularization and inhibit inflammation [16]. Recent studies suggest that the therapeutic effects exerted by PMSCs are likely due to the secretion of bioactive substances [6]. Lee et al. demonstrated that the exosomes secreted by MSCs have cytoprotective properties [17]. PMSCs express wide range of cytokines involved in wound healing, including IL-6 and IL-8, VEGF, angiogenin, PDGF, TGF-beta 2, and TIM-1 [18, 19]. When compared to BM-MSCs, AMSCs exhibited a different paracrine factor profile [20]. The pathway for cytokine secretion activation was also different in fetal PMSCs compared to maternal PMSCs when exposed to immunostimulatory factors [11]. Essentially, the secretion of PMSCs is a key factor of their therapeutic utility and should be taken into consideration for therapeutic applications. Immunomodulation of PMSCs Organ and cell transplantation approaches are often limited due to the graft-versus-host disease (GVHD). One of the most attractive properties of MSCs is their immunomodulatory effect that may overcome GVHD [21]. Their immunosuppression qualities are linked to inhibition of T cell and other immune cells response [22], and administration of BM-MSCs was correlated with a decrease or even absence of GVHD after transplantation of bone marrow and liver [23, 24]. The unique ability of the placenta to maintain fetomaternal tolerance indicates the immunomodulatory potential of PMSCs, and recently, PMSCs were shown to have regenerative immunomodulatory capabilities similar to BM-MSC, including T cell proliferation and cytokine secretion inhibition [25]. Moreover, PMSCs are more efficient when used for expansion of CD34? HSCs from umbilical cord blood, which are regularly used in cell replacement therapies in blood and immune system diseases. As a result, PMSCs

11

can serve as an alternative to bone marrow stromal cells for use in the expansion of blood progenitor cells in vitro and as support for hematopoiesis in vivo. In another study, PMSCs were shown to diminish chronic lung allograft rejection in a murine model [26], and tissues treated with PMSC as well as PMSC-conditioned medium demonstrated increase epithelial progenitor cells and reduction of neutrophils and activated T cells. These results suggest a possible role for PMSCs in the reduction of chronic rejection followed by organ transplantation, and interestingly, chorionic plate-derived mesenchymal stem cells were also shown to slow the formation of fibrosis and CCl4-induced cirrhosis in mice [27]. Thus, there is significant evidence for PMSCs as potent anti-inflammatory and immunomodulatory agents.

PMSCs from Different Placenta Regions The placenta consists of four anatomic regions: amniotic epithelial, amniotic mesenchymal, chorionic mesenchymal, and chorionic trophoblastic [28], and the decidua is the section of uterine lining that forms the maternal part of the placenta during pregnancy. Due to the high level of complexity of the human placenta and the limited scope of this review, only the amnion, chorion, and decidua will be discussed as sources of stem cells in this review paper. AEC, amniotic mesenchymal stromal cells (AMSCs), chorionic mesenchymal stromal cells (CMSCs), and chorionic trophoblastic cells (CTCs) can be isolated from these regions [28]. Other placental sources such as umbilical cord, Wharton’s Jelly, cord blood, and amniotic fluid will not be included in this review. The human fetal membranes—the amnion and chorion—envelop the fetus and its surrounding amniotic fluid, forming a highly specialized interface between mother and fetus that carries out important functions during pregnancy [29]. The amnion is a thin, avascular membrane composed of a single layer of epithelial cells and a layer of mesenchymal stromal cells, and these two cell layers are the sources of human amniotic multipotent epithelial cells (hAECs) and amniotic MSCs (AMSCs) [28]. The chorion consists of a chorionic mesenchymal stromal layer covering a chorionic trophoblast cell layer, which are the sources of chorionic MSCs (CMSCs) and hCTCs, respectively [28]. The chorionic plate is a multilayered structure that consists of the amniotic membrane and the chorion, and the chorionic plate extends from the chorionic disc to enclose the fetus in the amniotic cavity. Chorionic villi originate from the chorionic plate and anchor the placenta through the trophoblast of the basal plate and maternal endometrium [28]. As the fetus grows, the fetal membranes enlarge, such that the chorion pushes into—and

123

12

subsequently adheres to—the overlying decidua parietalis, which is of maternal origin [29]. The chorion with attached decidua parietalis is called the choriodecidua. From the maternal side, protrusions of the basal plate within the chorionic villi produce the placental septa, which divide the parenchyma into irregular cotyledons. Table 1 presents a brief overview of recent studies that have used term PMSCs from amnion, chorion, and decidua for experiments in regenerative medicine. Preterm PMSCs, hAECs, and CTCs will be addressed later in this review paper. Effect of Gestational Age on PMSC Properties Preterm PMSCs present a potential source of autologous MSCs for cell therapy and regenerative medicine. In their review chapter ‘‘Fetal Mesenchymal Stem Cells are More Primitive than Adult Mesenchymal Stem Cells’’ [42], Go¨therstro¨m et al. present a brief overview of prior experiments involving first- and second-trimester fetal tissues. Human MSCs have been isolated from several fetal tissues, including first trimester blood, bone marrow, liver, and placenta [43–45] and from second trimester blood, bone marrow, liver, lung, spleen, pancreas, kidney, brain, and amniotic fluid [46–51]. Collection of fetal somatic tissues during the first trimester is technically challenging, but the placenta is a larger and easier tissue to separate than other fetal tissues and is a more realistic candidate for cell banking. However, acquiring fetal tissue or preterm placenta can require termination of the pregnancy, which makes them ethically contentious as a cell source for autologous applications [52••]. This ethical concern contrasts sharply to term placenta, which is commonly discarded after term delivery and therefore lacks many of the ethical concerns of preterm placenta. As a result, the ability to research preterm PMSCs is limited compared to term PMSCs. However, preterm PMSCs can also be isolated in ongoing pregnancies from surplus tissues obtained during routine prenatal diagnostic procedures, such as chorionic villous sampling [45, 53, 54] and amniocentesis [55–57]. Table 2 presents a brief overview of recent studies that shows the potential of PMSCs for regenerative medicine. The results demonstrate that PMSCs have varied phenotypic characteristics depending on the developmental stage of the placenta, and these findings suggest that early gestational placentas are closer to the embryonic stage than the late full term placentas are [52••]. Current results suggest that preterm PMSCs may exhibit greater growth capacity [58], proliferation potential [58, 59], differentiation potential [60], and stability for long-term cultivation [61•, 62] than term PMSCs. These results suggest significant potential for the use of preterm PMSCs for tissue engineering and regenerative therapy [9, 52••]; however,

123

Curr Pathobiol Rep (2015) 3:9–16

further studies and examination of ethical issues are needed.

Amniotic Epithelial Cells AEC from the placenta have been investigated due to their phenotypic similarity to embryonic stem cells and their ability to differentiate into all types of cells from three germ layers [14, 64]. Marongiu et al. demonstrated the transdifferentiation potential of human AECs into hepatocytes, and after transplantation of human AECs into SCID/ Beige mice, Marongiu et al. also examined the capacity of AECs to repopulate the liver and differentiate into functional hepatocytes [65]. In vitro hepatocyte differentiation of hAECs showed expression of hepatocyte markers similar to fetal hepatocytes, and after a 3-week differentiation protocol using extracellular matrix (ECM) as a substrate for differentiation, there was an up-regulation of liver genes, such as Albumin, A1AT, CYP3A4, 3A7, 1A2, 2B6, and the Asialoglycoprotein receptor 1 (ASGPR1). In vivo, naive human AECs transplanted into SCID/Beige mice pretreated with retrorsine (RS) expressed mature liver genes 6 months after transplantation. This study suggests that a suitable microenvironment for hepatocyte differentiation is needed for high-level long-term engraftment of transdifferentiated hAECs. To improve survivability of AECs and prevent the effects of immunogenicity, Vaghjiani et al. encapsulated AECs in barium alginate microcapsules [66]. Examination of phenotype and function of AECs after a four-week differentiation of AECs demonstrated functional hepatocytes exhibiting the ability to synthesize urea and metabolize drugs via cytochrome P450. After encapsulation, cells remained viable, and liver functions were improved. The use of alginate capsules may be a viable approach for avoidance of immune rejection. However, cell-to-cell contact and exchange of nutrients into the encapsulated cells are limitations to this approach.

Hematopoietic Stem Cells (HSCs) HSCs are capable of generating cells that populate the blood and immune system, and HSCs are among the most extensively studied type of stem cell for therapy. In adults, HSCs reside in the bone marrow, and they are effectively the driving component behind efficacy of bone marrow transplantation. In the fetal environment, however, hematopoiesis (the process of forming blood cells and components) occurs in several regions prior to the bone marrow. Recently, researchers have discovered that the placenta is a hematopoietic organ capable of

Curr Pathobiol Rep (2015) 3:9–16

13

Table 1 Recent studies using term PMSCs in regenerative medicine Source

Year

Author

Investigation

Experimental findings

Amnion

2010

Chang et al. [30]

Identified MSCs from the amniotic membrane mesoderm

AMSCs demonstrated MSC tri-lineage potential and neuronal differentiation in vitro

Amnion

2011

Sus¸ man et al. [31]

Evaluated the effect of co-culture technique on exocrine pancreatic cells and placental stem cells

AMSCs changed in phenotype from mesenchymal to epithelial-like cells and expressed pancreatic amylase

Amnion

2012

Warrier et al. [32]

Investigated the potential of AMSCs for wound healing and blood vessel formation

AMSCs have pro-angiogenic, anti-fibrotic, and low immunogenicity properties

Amnion

2013

Avanzi et al. [33]

Investigated the susceptibility of AMSCs to human herpes virus infection

AMSCs were fully permissive to infection from several herpes viruses

Amnion

2014

Carbone et al. [34]

Investigated the regenerative and reparative potential of AMSCs for treatment of lung diseases

AMSC reduced pulmonary fibrosis and inflammation in an in vivo bleomycininduced lung injury model

Amnion, chorionic villi, chorionic plate

2012

Lee et al. [35]

Compared the hepatogenic differentiation potential of PMSCs with AMSCs, BMMSCs, and UCB-MSCs

PMSCs showed higher hepatogenic differentiation efficiency than AMSCs, BM-MSCs, and UCB-MSCs

Amnion, chorion

2014

Kinzer et al. [36]

Evaluated the effects of PMSCs on neovascularization in a mouse model

PMSC/EC co-culture promoted neovascularization in the mouse model

Chorionic villi

2010

Fazekasova et al. [37]

Compared immunomodulatory properties of PD-MSCs to BM-MSCs

BM-MSCs were more immunomodulatory than PD-MSCs

Chorion

2011

Tran et al. [38]

Isolated CD349? and CD349- fractions from PMSCS and examined effect on fractured bone and blood flow in ischemic regions of mouse models

CD349? and CD349- fractions facilitated new bone calcification in fractured femurs

Chorionic plate

2012

Lee et al. [21]

Compared immunomodulatory function of CP-MSCs with that of BM-MSCs and ADMSCs

CP-MSCs secrete cytokines differently than BM-MSCs and AMSCS, and CP-MSCs may have advantages in regenerative capacity and immunomodulation

Decidua basalis

2012

Fan et al. [39]

Explored TEBs constructed by PMSCs and their effect for the repair of osteoperiosteal defects

TEBs constructed by both PMSCs and BMSCs repaired the osteoperiosteal defects in a ‘multipoint’ manner

Decidua Parietalis

2012

Castrechini et al. [29]

To determine if decidua parietalis is an MSC niche

Decidua parietalis can serve as a niche for vascular stem cells

Decidua parietalis

2013

Fukusumi et al. [40]

Examined PCM from DP-MSCs for longterm stable maintenance of IPSCs

PCM from DM-MSCs can be used for the feeder-cell-free generation and long-term stable maintenance of IPSCs

Decidua parietalis

2011

Kanematsu et al. [41]

Compared the cellular properties of DPMSCs and adult BM-MSCs

DP-MSCs showed high proliferation capacity, differentiated well into chondrocytes, and moderately well into adipocytes. DP-MSCs hardly differentiated into osteoblasts

Mixed

2014

Zhao et al. [26]

Investigated effect of PMSCs in reducing development of bronchiolitis obliterans in a murine heterotopic tracheal transplant model

PMSCs are protective against the development of bronchiolitis obliterans in a heterotopic tracheal transplant model

AMSC amnion-derived mesenchymal stem cells, BM-MSCs bone marrow-derived mesenchymal stem cells, CP-MSCs chorionic plate-derived mesenchymal stem cells, DP-MSCs decidua parietalis-derived mesenchymal stem cells, EC endothelial cells, IPSCs induced pluripotent stem cells, PCM pericellular matrix, PMSCs placenta-derived mesenchymal stem cells, TEB tissue-engineered bone, UCB-MSCs umbilical cord blood-derived mesenchymal stem cells

supporting hematopoiesis and certain key points in development [67, 68•]. While the placenta has not generally been known as a source for HSCs to be used in therapy, the new identification of these cells within the placenta may drive future research into the isolation and

potential expansion of HSCs from the placenta. One study identified CD34? CD45? cells that had clonogenic potential and were capable of generating myeloid and erythroid progenitors, as well as natural killer cells and B cells in vitro [68•]. The researchers noted, however, that

123

14

Curr Pathobiol Rep (2015) 3:9–16

Table 2 Recent publications using preterm PMSCs in regenerative medicine Year

Author

Tissue

Investigation

Experimental findings

2010

Sung et al. [58]

First-trimester (8–12 weeks) and third-trimester (38–40 weeks) chorionic plate amnion

Evaluated the proliferation capacity, phenotypic expression, mesoderm differentiation, and expression of pluripotency stem cell markers between preterm and term MSCs

Preterm PMSCs had better growth capacity and proliferation potential with the expression of pluripotency markers

2011

Poloni et al. [61•]

2012

Jones et al. [52••]

First-trimester (11–13 weeks) chorionic villi Preterm (8–12 weeks) and term chorionic villous

Evaluated susceptibility of PMSCs to malignant transformation after longterm culture Investigated whether preterm or term placental chorionic stem cells are superior for cell therapy and tissue engineering

Fetal MSCs can be expanded long term with no increase in telomerase activity and no evidence of genetic changes Preterm PMSCs have smaller size, faster kinetics, and more favorable genetic markers for use in regenerative medicine

2013

Park et al. [63]

First-trimester and thirdtrimester placenta

Compared the pluripotency of firsttrimester and third-trimester placenta

Preterm PMSCs have 2–11-fold higher expression of pluripotency-coupled genes than term PMSCs, implying greater differentiation potential

2013

Roselli et al. [62]

First-trimester (11–12 weeks) chorionic villi

Determined the genetic stability of chorionic villi in long-term culture

CV-MSCs are genetically stable in longterm cultures at least up to passage 10

2014

Di bernardo et al. [9]

Preterm (34–39 weeks) chorion

Investigated the effects of preterm PMSCs as a potential therapy for neonatal respiratory failure secondary to pulmonary hypoplasia

Preterm PMSCs enhanced perinatal lung growth

2014

Youssef et al. [59]

Preterm (10–13 weeks) and term (37–42 weeks) PMSCs

Investigated the impact of varying concentrations of oxygen tension and IGF-I on preterm and term PMSC multipotency

Preterm PMSCs had greater proliferation response to IGF-I, were enhanced by low-oxygen tension, and were more multipotent than term PMSCs

AMSC amnion-derived mesenchymal stem cells, BM-MSCs bone marrow-derived mesenchymal stem cells, CP-MSCs chorionic plate-derived mesenchymal stem cells, CV-MSC chorionic villi-derived mesenchymal stem cells, DP-MSCs decidua parietalis-derived mesenchymal stem cells), IPSCs induced pluripotent stem cells, PCM pericellular matrix, PMSCs placenta-derived mesenchymal stem cells, UCB-MSCs umbilical cord blood-derived mesenchymal stem cells

the presence of these cells peaked within 5–8 weeks gestation, making them a difficult source to obtain for cell therapy. Still, more research is needed to determine if HSCs can be obtained from the placenta later in gestation or even at term in order for the placenta to remain a viable option as a source of HSCs for future therapy.

derived neural stem cells were able to regenerate the dopaminergic nigrostriatal pathway in both acute and chronic Parkinson’s disease rat models. While this study indicates the great potential for these cells to be utilized for regenerative medicine, much work still needs to be done in order to understand the potential benefits of using trophoblasts as compared to other more commonly used cell types such as MSCs.

Trophoblast Stem Cells Trophoblasts are a cell population entirely unique to the placenta, and they arise from the trophectoderm layer of the blastocyst early in development and facilitate embryo implantation and formation of the placenta. They have been widely studied for their importance in the developmental process, but it may be possible to utilize isolated trophoblasts and TSCs for cell therapy. Lee et al. demonstrated the capability of TSCs obtained from ectopic pregnancies to differentiate into neural stem cells [69], and the study found that after transplantation, these trophoblast-

123

Conclusion The placenta is a unique organ of fetomaternal origin that can serve as a source of multiple types of stem cells for autologous and allogeneic cell transplantation. PMSCs, AECs, HSCs, and TSCs can be isolated from different anatomic regions of the placenta, and these stem cells have great potential, due to their expansion rate, differentiation capability, secretion, and immunomodulation functions. However, more in vivo disease model research is needed to

Curr Pathobiol Rep (2015) 3:9–16

verify the safety and efficacy of placenta-derived stem cells for application in regenerative medicine. Compliance with Ethics Guidelines Conflict of Interest Jason A. Meierhenry, Volodymyr Ryzhuk, Maricel G. Miguelino, Lee Lankford, Jerry S. Powell, Diana Farmer and Aijun Wang declare that they have no conflict of interest. Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.

References Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance 1. Ryan JM et al (2013) Unravelling the pluripotency paradox in fetal and placental mesenchymal stem cells: Oct-4 expression and the case of The Emperor’s New Clothes. Stem Cell Rev 9(4):408–421 2. Dominici M et al (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8(4):315–317 3. Friedenstein AJ et al (1974) Precursors for fibroblasts in different populations of hematopoietic cells as detected by the in vitro colony assay method. Exp Hematol 2(2):83–92 4. Pontikoglou C et al (2011) Bone marrow mesenchymal stem cells: biological properties and their role in hematopoiesis and hematopoietic stem cell transplantation. Stem Cell Rev 7(3):569–589 5. Huang HI (2007) Isolation of human placenta-derived multipotent cells and in vitro differentiation into hepatocyte-like cells. Curr Protoc Stem Cell Biol. Chapter 1, p Unit 1E.1 6. Caruso M, Evangelista M, Parolini O (2012) Human term placental cells: phenotype, properties and new avenues in regenerative medicine. Int J Mol Cell Med 1(2):64–74 7. Parolini O, Caruso M (2011) Review: preclinical studies on placenta-derived cells and amniotic membrane: an update. Placenta 32(Suppl 2):S186–S195 8. Makhoul G, Chiu RCJ, Cecere R (2013) Placental mesenchymal stem cells: a unique source for cellular cardiomyoplasty. Ann Thorac Surg 95(5):1827–1833 9. Di Bernardo J et al (2014) Paracrine regulation of fetal lung morphogenesis using human placenta-derived mesenchymal stromal cells. J Surg Res 190(1):255–263 10. Barlow S et al (2008) Comparison of human placenta- and bone marrow-derived multipotent mesenchymal stem cells. Stem Cells Dev 17(6):1095–1107 11. Zhu Y et al (2014) Placental mesenchymal stem cells of fetal and maternal origins demonstrate different therapeutic potentials. Stem Cell Res Ther 5(2):48 12. Meraviglia V et al (2012) Human chorionic villus mesenchymal stromal cells reveal strong endothelial conversion properties. Differentiation 83(5):260–270 13. De Coppi P et al (2007) Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 25(1):100–106 14. Miki T et al (2005) Stem cell characteristics of amniotic epithelial cells. Stem Cells 23(10):1549–1559

15 15. Soncini M et al (2007) Isolation and characterization of mesenchymal cells from human fetal membranes. J Tissue Eng Regen Med 1(4):296–305 16. James JL et al (2014) Can we fix it? Evaluating the potential of placental stem cells for the treatment of pregnancy disorders. Placenta 35(2):77–84 17. Lee C et al (2012) Exosomes mediate the cytoprotective action of mesenchymal stromal cells on hypoxia-induced pulmonary hypertension. Circulation 126(22):2601–2611 18. Liu SH et al (2010) Paracrine factors from human placental multipotent mesenchymal stromal cells protect endothelium from oxidative injury via STAT3 and manganese superoxide dismutase activation. Biol Reprod 82(5):905–913 19. Steed DL et al (2008) Amnion-derived cellular cytokine solution: a physiological combination of cytokines for wound healing. Eplasty 8:e18 20. Wegmeyer H et al (2013) Mesenchymal stromal cell characteristics vary depending on their origin. Stem Cells Dev 22(19):2606–2618 21. Lee JM et al (2012) Comparison of immunomodulatory effects of placenta mesenchymal stem cells with bone marrow and adipose mesenchymal stem cells. Int Immunopharmacol 13(2):219–224 22. Caplan AI (2009) Why are MSCs therapeutic? New data: new insight. J Pathol 217(2):318–324 23. Dazzi F, Marelli-Berg FM (2008) Mesenchymal stem cells for graft-versus-host disease: close encounters with T cells. Eur J Immunol 38(6):1479–1482 24. Xia X et al (2012) Mesenchymal stem cells administered after liver transplantation prevent acute graft-versus-host disease in rats. Liver Transplant 18(6):696–706 25. Luan X et al (2013) Human placenta-derived mesenchymal stem cells suppress T cell proliferation and support the culture expansion of cord blood CD34(?) cells: a comparison with human bone marrow-derived mesenchymal stem cells. Tissue Cell 45(1):32–38 26. Zhao Y et al (2014) Treatment with placenta-derived mesenchymal stem cells mitigates development of bronchiolitis obliterans in a murine model. J Thorac Cardiovasc Surg 147(5): 1668–1677.e5 27. Zhang D, Jiang M, Miao D (2011) Transplanted human amniotic membrane-derived mesenchymal stem cells ameliorate carbon tetrachloride-induced liver cirrhosis in mouse. PLoS ONE 6(2):e16789 28. Parolini O et al (2008) Concise review: isolation and characterization of cells from human term placenta: outcome of the first international workshop on placenta derived stem cells. Stem Cells 26(2):300–311 29. Castrechini NM et al (2012) Decidua parietalis-derived mesenchymal stromal cells reside in a vascular niche within the choriodecidua. Reprod Sci 19(12):1302–1314 30. Chang YJ et al (2010) Isolation of mesenchymal stem cells with neurogenic potential from the mesoderm of the amniotic membrane. Cells Tissues Organs 192(2):93–105 31. Sus¸ man S et al (2011) Pancreatic exocrine adult cells and placental stem cells co-culture. Working together is always the best way to go. Rom J Morphol Embryol 52(3 Suppl): 999–1004 32. Warrier S, Haridas N, Bhonde R (2012) Inherent propensity of amnion-derived mesenchymal stem cells towards endothelial lineage: vascularization from an avascular tissue. Placenta 33(10):850–858 33. Avanzi S et al (2013) Susceptibility of human placenta derived mesenchymal stromal/stem cells to human herpesviruses infection. PLoS ONE 8(8):e71412 34. Carbone A et al (2014) Human amnion-derived cells: prospects for the treatment of lung diseases. Curr Stem Cell Res Ther 9(4):297–305

123

16 35. Lee HJ et al (2012) Comparison of in vitro hepatogenic differentiation potential between various placenta-derived stem cells and other adult stem cells as an alternative source of functional hepatocytes. Differentiation 84(3):223–231 36. Kinzer M et al (2014) Mesenchymal stromal cells from the human placenta promote neovascularization in a mouse model in vivo. Placenta 35(7):517–519 37. Fazekasova H et al (2011) Placenta-derived MSCs are partially immunogenic and less immunomodulatory than bone marrowderived MSCs. J Tissue Eng Regen Med 5(9):684–694 38. Tran TC et al (2011) Identification of human placenta-derived mesenchymal stem cells involved in re-endothelialization. J Cell Physiol 226(1):224–235 39. Fan ZX et al (2012) Placenta- versus bone-marrow-derived mesenchymal cells for the repair of segmental bone defects in a rabbit model. FEBS J 279(13):2455–2465 40. Fukusumi H et al (2013) Feeder-free generation and long-term culture of human induced pluripotent stem cells using pericellular matrix of decidua derived mesenchymal cells. PLoS ONE 8(1):e55226 41. Kanematsu D et al (2011) Isolation and cellular properties of mesenchymal cells derived from the decidua of human term placenta. Differentiation 82(2):77–88 42. Go¨therstro¨m C et al (2007) Fetal mesenchymal stem cells are more primitive than adult mesenchymal stem cells. In: Habib N et al (eds) Stem cell repair and regeneration, vol 2. Imperial College Press, London, pp 17–34 43. Campagnoli C et al (2001) Identification of mesenchymal stem/ progenitor cells in human first-trimester fetal blood, liver, and bone marrow. Blood 98:2396–2402 44. Gotherstrom C et al (2003) Immunomodulatory effects of human foetal liver-derived mesenchymal stem cells. Bone Marrow Transplant 32:265–272 45. Portmann-Lanz CB et al (2006) Placental mesenchymal stem cells as potential autologous graft for pre- and perinatal neuroregeneration. Am J Obstet Gynecol 194:664–673 46. Almeida-Porada G et al (2002) Differentiative potential of human metanephric mesenchymal cells. Exp Hematol 30:1454–1462 47. In’t Anker PS et al (2002) Mesenchymal stem cells in human second-trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential. Haematologica 88:845–852 48. In’t Anker PS et al (2003) Amniotic fluid as a novel source of mesenchymal stem cells for therapeutic transplantation. Blood 102:1548–1549 49. Hu Y et al (2003) Isolation and identification of mesenchymal stem cells from human fetal pancreas. J Lab Clin Med 141:342–349 50. Airey JA et al (2004) Human mesenchymal stem cells form Purkinje fibers in fetal sheep heart. Circulation 109:1401–1407 51. Yu M et al (2004) Mid-trimester fetal blood-derived adherent cells share characteristics similar to mesenchymal stem cells but fullterm umbilical cord blood does not. Br J Haematol 124:666–675 52. •• Jones GN et al (2012) Ontological differences in first compared to third trimester human fetal placental chorionic stem cells. PLoS ONE 7(9):e43395. This study demonstrated that first trimester PMSCs have more primitive and curative characteristics than third trimester PMSCs in vitro and in vivo. It provides insight into the ontogeny of the stemness phenotype during fetal development and suggests that the more primitive characteristics of early gestation fetal chorionic stem cells may be translationally advantageous compared to those of late gestation 53. Poloni A, Rosini V, Mondini E et al (2008) Characterization and expansion of mesenchymal progenitor cells from first trimester chorionic villi of human placenta. Cytotherapy 10:690–697

123

Curr Pathobiol Rep (2015) 3:9–16 54. Spitalieri P, Cortese G, Pietropolli A et al (2009) Identification of multipotent cytotrophoblast cells from human first trimester chorionic villi. Cloning Stem Cells 11:535–556 55. De Coppi P, Bartsch G Jr, Siddiqui MM, Xu T, Santos CC et al (2007) Isolation of amniotic stem cell lines with potential for therapy. Nat Biotechnol 25:100–106 56. Tsai MS, Lee JL, Chang YJ et al (2004) Isolation of human multipotent mesenchymal stem cells from second-trimester amniotic fluid using a novel two stage culture protocol. Hum Reprod 19:1450–1456 57. Prusa AR, Marton E, Rosner M et al (2003) Oct-4-expressing cells in human amniotic fluid: a new source for stem cell research? Hum Reprod 18:1489–1493 58. Sung HJ et al (2010) Stemness evaluation of mesenchymal stem cells from placentas according to developmental stage: comparison to those from adult bone marrow. J Korean Med Sci 25(10):1418–1426 59. Youssef A, Iosef C, Han VK (2014) Low-oxygen tension and IGF-I promote proliferation and multipotency of placental mesenchymal stem cells (PMSCs) from different gestations via distinct signaling pathways. Endocrinology 155(4):1386–1397 60. Han NR et al (2013) Generation of priming mesenchymal stem cells with enhanced potential to differentiate into specific cell lineages using extracellular matrix proteins. Biochem Biophys Res Commun 436(3):413–417 61. • Poloni A et al (2011) Human mesenchymal stem cells from chorionic villi and amniotic fluid are not susceptible to transformation after extensive in vitro expansion. Cell Transplant 20(5):643–654. This study demonstrated that human mesenchymal progenitor cells derived from chorionic villi (CV) and amniotic fluid (AF) isolated during the first and second trimesters, respectively, preserve a constant expression level of p53 and a normal karyotype throughout long-term expansion. These findings suggest the safety of fetal MSCs for cell therapy and regenerative medicine 63. Park S et al (2013) Comparison of human first and third trimester placental mesenchymal stem cell. Cell Biol Int 37(3):242–249 62. Roselli EA et al (2013) Fetal mesenchymal stromal cells from cryopreserved human chorionic villi: cytogenetic and molecular analysis of genome stability in long-term cultures. Cytotherapy 15(11):1340–1351 64. Akle CA et al (1981) Immunogenicity of human amniotic epithelial cells after transplantation into volunteers. Lancet 2(8254): 1003–1005 65. Marongiu F et al (2011) Hepatic differentiation of amniotic epithelial cells. Hepatology 53(5):1719–1729 66. Vaghjiani V et al (2014) Hepatocyte-like cells derived from human amniotic epithelial cells can be encapsulated without loss of viability or function in vitro. Stem Cells Dev 23(8):866–876 67. Robin C et al (2009) Human placenta is a potent hematopoietic niche containing hematopoietic stem and progenitor cells throughout development. Cell Stem Cell 5(4):385–395 68. • Barcena A et al (2009) A new role for the human placenta as a hematopoietic site throughout gestation. Reprod Sci 16(2): 178–187. This study shows that human placenta contains primitive multipotent and intermediate progenitors and that the cell number and density change over gestation. The findings suggest that human placenta is potentially an important hematopoietic organ, opening the possibility of banking placental hematopoietic stem cells along with cord blood for transplantation 69. Lee TT et al (2012) Ectopic pregnancy-derived human trophoblastic stem cells regenerate dopaminergic nigrostriatal pathway to treat parkinsonian rats. PLoS ONE 7(12):e52491