Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 1.
Published in final edited form as: Arterioscler Thromb Vasc Biol. 2010 May 7;30(6):1110–1117. doi: 10.1161/ATVBAHA.109.191601

Vascular Potential of Human Pluripotent Stem Cells

Ionela Iacobas 1,3,5, Archana Vats 1,3,4, Karen K Hirschi 1,2,3,4,6
PMCID: PMC2896478  NIHMSID: NIHMS203360  PMID: 20453170

Abstract

Cardiovascular disease is the number one cause of death and disability in the US. Understanding the biological activity of stem and progenitor cells, and their ability to contribute to the repair, regeneration and remodeling of the heart and blood vessels affected by pathologic processes is an essential part of the paradigm in enabling us to achieve a reduction in related deaths. Both human embryonic stem (ES) cells and induced pluripotent stem (iPS) cells are promising sources of cells for clinical cardiovascular therapies. Additional in vitro studies are needed, however, to understand their relative phenotypes and molecular regulation toward cardiovascular cell fates. Further studies in translational animal models are also needed to gain insights into the potential and function of both human ES- and iPS-derived cardiovascular cells, and enable translation from experimental and pre-clinical studies to human trials.

Clinical Need to Control Blood Vessel Formation

The vasculature is a ubiquitously distributed organ system that functions to provide oxygen and nutrients, as well as remove metabolic waste products, throughout the entire body. Disruption of blood vessel formation and/or function plays a central role in the progression of many prevalent disease processes. Thus, the clinical need for controlled blood vessel formation (promotion or suppression) plays a central role in many therapeutic approaches.

In the 1970s, Judah Folkman championed a vigorous campaign to suppress aberrant blood vessel formation for the treatment of angiogenesis-dependent diseases1 (i.e. cancer, infantile hemangiomas2, peptic ulcers3, ocular neovascularization4, rheumatoid arthritis5 and atherosclerosis6,7,8). Since then, more than ten anti-angiogenic agents have entered clinical trials or have been approved for human use.

Equally challenging has been the effort to develop pro-angiogenic therapies for tissues suffering from chronic or acute ischemia, for tissues engineered ex vivo, and for the vascularization of implanted tissue grafts. Many groups have tried to stimulate endogenous neovascularization using various strategies including systemic administration of pro-angiogenic factors, mobilization of bone marrow-derived cells, and transplantation of putative vascular progenitor cells; all with promising, but still limited, results.

In conjunction with the need to optimize strategies to promote neovascularization in vivo, is the need to continuously identify, and evaluate the use of, human stem/progenitor cell types with potential to serve as vascular cell precursors. Since the discovery of human stem cells with pluripotent properties, including human embryonic stem (ES) cells9 and human induced pluripotent stem (iPS) cells10, there has been increasing interest in characterizing these cell types and controlling their differentiation towards specific cellular lineages. Herein, we will provide an overview of the vascular potential of human pluripotent stem cells, and endothelial cell differentiation there from.

Vasculogenesis

In situ differentiation of endothelial cells from multipotent progenitor cells, and the formation of endothelial tube networks, is referred to as vasculogenesis. The subsequent stages of blood vessel formation, including network remodeling and recruitment of surrounding mural cells (pericytes and smooth muscle cells), is referred to as angiogenesis. In mammals, extraembryonic vasculogenesis precedes intraembryonic vascular development and is initiated shortly after gastrulation, as cells from the epiblast migrate through the primitive streak and organize into the mesodermal germ layer11,12. While mesodermal cells are the most widely recognized source of embryonic endothelial precursors13,14,15, neural progenitors have demonstrated the ability to differentiate into endothelial cells in certain conditions16,17. Nonetheless, the first endothelial cells are derived from mesodermal precursors in the extraembryonic yolk sac in structures referred to as blood islands, which consist of “primordial” (non-specialized) endothelial cells, as well as primitive erythroblasts18,19,20. Subsequent coalescence of blood islands contributes to the formation of a plexus of endothelial tubes that is then remodeled into a circulatory network that is invested by recruited mural cells.

Regulation of Endothelial Cell Development

Important insights into the regulation of endothelial cell differentiation have been gained using multiple development models including avian, zebrafish and mouse embryos. Studies, particularly in the mouse model system, have revealed soluble factors and transcriptional regulators involved in endothelial cell development in vivo, as discussed below and summarized in Table 1.

Table 1.

Factors Implicated in Murine Endothelial Cell Development

Soluble factors Transcription factors
Ihh - Expressed at E6.5 in visceral endoderm21,109,110
- Required for vasculogenesis
Forkhead Family - Expressed in the splanchnic mesoderm
- Important in the arterial and lymphatic proliferation
BMP4 - Expressed prior to gastrulation
- Required for mesoderm formation28,111113
Klf Family - Expressed at E7
- Required for vessel stabilization116
bFGF - FGFR1 expressed on ectodermal cells at the egg cylinder stage and on migrating mesodermal cells114
- Controls mesodermal differentiation.
Ets Family - ER71 expressed at E7.5
- Required for induction of Flk1 expression37,117
VEGFA/Flk-1 - Expressed at E4 in the primitive endoderm, and by E7 expression is restricted to the extraembryonic endoderm and mesoderm115
- Required for migration and proliferation of endothelial cells
Scl/tal-1 - Expressed at E7.5 in mesodermal progenitors118
- Regulates Flk-1 and VE-cadherin expression
GATA 1/2 - Signals downstream of BMP4 to induce Scl expression and endothelial differentiation
Vezf1 - Required for proper endothelial cell-cell junctions formation

Signaling pathways

In the developing mouse, it is suggested that during yolk sac vasculogenesis, visceral endoderm-derived soluble factors, such as Indian Hedgehog (Ihh)16,21, vascular endothelial growth factor (VEGF-A)22, and basic fibroblast growth factor (bFGF)23 promote endothelial cell development within the underlying mesoderm where their receptors, Ptc, VEGFR2/Flk1 and FGFR2, respectively, are localized16,2427. Other signaling molecules proposed to be downstream targets of Ihh and VEGF-A signaling, such as bone morphogenic protein 4 (BMP4)16,28, are similarly localized within the mesoderm. While all of these factors have been shown, individually, to be of importance in regulating murine blood vessel formation, the signaling hierarchy among them has not been clearly delineated. Whether similar signals, in a specific hierarchy, also regulate human endothelial cell development has only recently been investigated, and will be discussed in subsequent sections of this review.

Transcriptional regulators

Several transcription factor families have been implicated in endothelial cell development, and presumably function in conjunction with (upstream or downstream) the soluble effectors discussed above.

Ets Transcription Factors

There are at least 19 Ets factors known to be expressed in human endothelial cells29; the most widely studied are Ets-1, Elf-1, Fli-1, Tel, Erg and ER71. All characterized endothelial enhancers and promoters contain multiple essential ETS binding sites, and ETS motifs are strongly associated with endothelial genes throughout the human genome30,31. Likely due to redundancy among Ets factors in endothelial cell development, germline deletion or mutation of the majority of individual Ets genes in either mouse or zebrafish model systems has resulted in little or no vascular phenotype or has caused defects only in later vascular remodeling, while vasculogenesis remained largely intact3236. One exception is Etv2 (ER71, Etsrp71) that appears to be essential for the development of endothelial and blood lineages in mouse. Expression of early vascular markers, such as VEGFR2/Flk-1, CD31/PECAM-1, and Tie-2 is almost completely abolished in the absence of Etv23739. Interestingly, while the majority of transcription factors have conserved functions among distinct species, Etv2 appears to be requried for myeloid lineage development in mice and zebrafish, but has no detectable effects on the expression of either myeloid or erythroid markers in Xenopus40.

Forkhead proteins

Although no Forkhead proteins are specifically expressed in endothelial cells or their known progenitors, targeted disruption of several family members (i.e. FoxO1, FoxF1, FoxC1/2) results in severe vascular phenotypes and embryonic lethality4144. FoxF1 is not expressed within endothelial cells of the differentiated embryonic vasculature, but is expressed earlier in the splanhnic mesoderm prior to endothelial cell specification and may regulate BMP signaling therein45. FoxC1 and C2 have important functions in arterial and lymphatic endothelial cell specialization and may be significant downstream effectors of Notch signaling in this process46,47

Kruppel-like factors (KLF)

Several KLF factors are known to be expressed in endothelial cells during vasculogenesis and early angiogenesis. KLF2 null mice die by embryonic day E14.5 due to hemorrhage resulting from lack of vessel stabilization and defective tunica media formation. In general, KLF family members appear to function within endothelial cells after initial specification and differentiation have occurred48.

GATA Factors

GATA factors have long been known to play important roles in blood cell development. GATA2, specifically, has been shown to be important not only for hematopoiesis49, but for endothelial cell development, as well50. In the mouse, GATA2 was shown to signal downstream of BMP4 within mesodermal progenitor cells to induce Scl/Tal-1 expression and differentiation into endothelial and hematopoietic cell types50.

Scl/Tal-1

Scl/Tal-1 is able to induce paraxial and non-axial mesodermal cells to produce hematopoietic and endothelial cells at the expense of somite and pronephric precursors, indicating that it could act to respecify mesodermal progenitors cells51. As mentioned above, Scl/Tal-1 appears to be modulated by BMP4 signaling52 and has been shown to regulate VEGFR2/Flk-1 and VE-cadherin expression in endothelial cells53.

Vezf1

Vezf1 is a zinc finger protein predominantly expressed in endothelial cells during early embryonic development54. In mice deficient for Vezf1, endothelial cells do not develop appropriate cell-cell junctions or deposit a normal extracellular matrix resulting in hemorrhage and embryonic death55.

Cell Systems to Study Endothelial Cell Differentiation

Although transcriptional regulators of endothelial cell differentiation have been implicated from embryonic studies, it is often difficult to delineate their precise, and potential interactive, role(s) in vivo due to functional redundancy with other family members or earlier functions within the embryo, including mesoderm formation. Many genes have also been deemed as essential for vascular development if their mutation or deletion resulted in embryonic lethality or caused a significant vascular phenotype in an animal model. However, every genetic manipulation affects not only the targeted gene but an array of functional pathways, whose coordinated function could be difficult to tease out in vivo. Thus, it is increasingly important that we take advantage of in vitro cell culture systems to delineate specific molecular regulators of endothelial cell differentiation from embryonic and adult progenitors, as well as determine the relevance of regulatory pathways identified in animal studies to modulating human endothelial cell formation for clinical therapeutic applications.

Various stem and progenitor cell types, with differing properties and potentials, have been shown to exhibit vascular potential. Although totipotent cells that can form cells of all lineages including the extraembryonic tissue (i.e. mammalian zygotes and early blastomeres56) can presumably form vascular cells, they are not typically used for vascular cell differentiation studies. In contrast, pluripotent cells such as ES9 and iPS57,58 cells, that have the ability to differentiate into all cell types of the body but not extraembryonic tissues (i.e. placenta), have been generated from human, as well as mouse, embryos and somatic cells, respectively. They propagate well in vitro, and have been adopted by many labs for the study of self-renewal and differentiation, including the differentiation of vascular cells. Multipotent cells, that are thought to be relatively lineage-restricted, such as those isolated from adult blood and blood vessels, also exhibit vascular potential; however, have proved less useful for molecular regulation studies due to heterogeneity and limited cell number. Thus, our review will specifically focus on the vascular potential of human pluripotent stem cells that serve as effective model systems for the study of early molecular events leading to endothelial cell development.

Human Pluripotent Stem Cells

Human ES cells

Since they were first isolated, there have been numerous reports demonstrating the differentiation potential of human ES cells into various derivatives of all three germ layers. Examples include ectodermal cells such as oligodendrocytes and neuroectoderm59,60, mesodermal derivatives including neutrophils and cardiomyocytes61,62 and endodermal cell types such as hepatocytes and pancreatic cells63,64. Along with these cell types, it has been well documented that human ES cells can generate both endothelial and hematopoietic cells6569. It has been a particular challenge, however, to identify markers specific to vascular endothelium that are not overlapping with hematopoietic cells that develop in parallel. Thus, when studying the differentiation of endothelial cells in vitro, their phenotype is best defined by co-expression of multiple markers (i.e. VEGFR2/Flk-1, CD31/PECAM-1, VE-cadherin), lack of expression of blood cell markers (i.e. CD45), and demonstrated endothelial cell function (i.e. tube formation, eNOS production)7074.

Among the methods available for vascular differentiation from ES cells, the most widely used are the embryoid body (EB) formation method and co-culture on monolayers of OP9 cells, which are murine bone marrow stromal cells. EBs are formed by dissociating undifferentiated human ES cells and plating them onto nonadherent plates, then supplementing with cytokines to promote their vascular differentiation and/or proliferation65,66,75. The OP9 co-culture method was originally developed for hematopoietic differentiation of mouse ES cells76 and later adapted for human ES cells67. The protocol allows dissociated undifferentiated human ES cells to be plated directly on top of OP9 cells that have been growth arrested and serve as a feeder layer to promote endothelial cell differentiation. The OP9 co-culture method enables more robust (~5–10%) endothelial cell production77 compared to the EB method (~1–2%), and requires less cytokine and growth factor supplementation.

A method to maintain human ES cells in an undifferentiated state on a feeder-free layer (i.e. Matrigel) with conditioned medium prepared from mouse embryonic fibroblast cultures was established in 200178, and adapted for use with an alternative conditioned media containing cloned zebrafish bFGF79. Endothelial cell generation from human ES cells maintained in a feeder-free culture system was achieved by growing the human ES cells on collagen IV-coated dishes with the addition of VEGF-A +/− pituitary extracts66.

iPS cells

Recently, reprogramming of differentiated adult cells to a state of pluripotency has been achieved using both human and murine fibroblasts57,58. In groundbreaking work, two groups independently transduced genes encoding four transcriptional regulators (Nanog, Lin28, Oct4 and Sox257 or c-Myc, Klf4, Oct4 and Sox258) in adult fibroblasts to induce a pluripotent phenotype. The resulting induced pluripotent stem (iPS) cells exhibited functional and genetic properties similar to that of human ES cells80. More recent reports indicate that induced pluripotency can be enhanced by small molecules such as methylation inhibitors, and can be achieved with as few as two reprogramming factors depending on the cell types used81.

A shortcoming, with regard to clinical potential, of the initial reprogramming method was the use of retro- or lenti-viruses to transduce fibroblasts. These viruses are integrated into host chromosomes where they can cause insertional mutagenesis, interfere with gene transcription, and induce malignant transformation82. In the first report of germline competent mouse iPS cells, 20% of chimeric mice developed tumors that were attributable to the reactivation of the c-Myc proviral transgene that had integrated into the host cell genome83. Another group reported cancer-related deaths in 18 of 36 iPS chimeric mice84. Thus, several groups have sought alternatives to retroviral gene integration. Yamanaka’s group has transfected plasmids without using viruses to generate mouse iPS cells85. Two recent papers have shown that mouse and human iPS cells can be produced by piggyBac transposition with four genes in a single plasmid, thus significantly improving induction efficiency86,87. Other methods employed Cre-recombinase-excisable viruses88,89, non-integrating episomal vectors90, insertion of transducing proteins91, and replication-defective adenoviral vectors92.

The process of deriving iPS cells is still not standardized, and there can be variation in the cell lines generated, even when using the same cells and induction protocol. For example, two cell lines derived from same donor using the same four factors show significant differences in hematopoietic differentiation potential, thought to be due to distinct viral integration sites in the clones93. Other groups94, however, that generated iPS cells by transfection with Oct3/4, Sox2, Klf4 +/− c-Myc and compared four human iPS and three human ES cell lines found no obvious differences among the iPS cells lines, and no differences between iPS and ES cells, with regard to differentiation potential when subjected to the same endothelial-derivation protocol. Although the majority of the published studies, to date, emphasize the similarities between human ES and iPS cells, it is important to note that differences in potential do exist and likely reflect differences in regulatory pathways controlling endothelial cell development from these distinct types of human pluripotent stem cells.

Molecular Regulation of Endothelial Cell Differentiation from Pluripotent Stem Cells

Most of our current knowledge about the molecular regulation of endothelial cell differentiation, as summarized above, has come from animal models. Thus, the role of specific factors must be thoroughly investigated in human cell systems before applying such insights to clinical therapeutics. Importantly, there are significant differences between murine and human development that may limit the usefulness of the mouse as a model. Human embryos, for instance, have two phases of extraembryonic endoderm formation and limited reliance on the yolk sac circulation, while the mouse has one phase of extraembryonic endoderm generation and utilizes the yolk sac until birth74. There are also known regulatory differences; for example, in humans, TGFβ inhibits the expression of endodermal, endothelial and hematopoietic markers95, which contrasts with findings in the mouse where exogenous TGFβ (TGFβ or overexpression of TGFβ receptor 2, Tgfβr2) reduces the level of endodermal markers but increases endothelial marker expression.

In addition, there are known differences between human and mouse ES cells. For example, human ES cells express a number of distinct cell surface antigens, exhibit leukemia inhibitory factor independency, and have a relatively long doubling time95,96. Therefore, it would not be surprising if there were differences in the molecular regulation of endothelial cell differentiation from mouse and human ES cells, as well as differences in the regulation of human ES and iPS cells. Herein, we summarize what is known about the regulation of endothelial cell differentiation from both types of human stem cells.

Human ES cells

In early studies of the role of VEGF-A in endothelial cell generation from human ES cells, it was determined that addition of this factor to cultures increased the production of endothelial cells65,66. Hence, it was suggested that VEGF-A promotes endothelial cell differentiation from human ES cells. However, the differentiation process in these studies was actually induced by other means (i.e. EB formation), and CD31-expressing cells were subsequently isolated from dissociated EB and expanded in response to exogenous VEGF-A. In more recent studies of the role of VEGF-A, as well as bFGF, in human ES cells, we determined that neither factor induces endothelial cell differentiation77. That is, when either or both are added to undifferentiated human ES cells, there is no increase in the expression of endothelial-specific genes or proteins. Although this does not exclude a role for either factor in later stages of endothelial cell development, such as survival or proliferation, neither factor appears to regulate the initial commitment to an endothelial cell lineage, which is consistent with studies in mouse ES cells, as previously reviewed97.

In contrast, we found that other factors such as Ihh and BMP do play an inductive role in the process of endothelial cell differentiation from human ES cells77. Ihh is expressed in the murine yolk sac visceral endoderm as early as 6.5dpc21,98. Although the specific cellular role for Ihh in murine vascular development is not defined, Ihh-null mutants are embryonic lethal, exhibit impaired yolk sac vasculogenesis and vascular remodeling, and the yolk sacs have fewer endothelial cells99. In studies in human ES cells, we found that exogenous Ihh increases the expression of BMP4, VEGF-A, and VEGFR2/Flk1, as well as generation of differentiated endothelial cells; conversely, inhibition of hedgehog signaling suppresses formation of endothelial cells66,68,100. Furthermore, inhibitors of BMP signaling abolish the Ihh-mediated effects, indicating that BMP factors likely signal downstream of Ihh to modulate human endothelial cell development. Consistent with this idea, addition of rhBMP4 to human ES cells, in conjunction with hedgehog inhibition, rescues endothelial cell formation to control levels. Collectively, these studies reveal that Ihh signals via the BMP pathway to promote endothelial cell differentiation from human ES cells77. Interestingly, a similar regulatory pathway has been defined for mouse ES cells, in which Ihh and BMP factors are needed to induce endothelial cell differentiation there from, and VEGF-A is necessary thereafter for endothelial cell expansion in culture97. Clearly, our understanding of the molecular regulation of endothelial cell differentiation from human ES cells is still very limited and more studies are needed to further dissect the regulatory pathways and factors involved.

iPS cells

The elements involved in the vascular differentiation of iPS cells have just started to be unraveled. The differentiation of human iPS cells toward an endothelial cell lineage can be induced using the same protocols used for the derivation of endothelial cells from human ES cells (i.e. EB and co-culture with OP9 cells), and the endothelial cell yield from human ES and iPS cells appears to be similar94. When VEGFR2/Flk-1- and VE-cadherin-expressing cells are sorted from iPS cell cultures and replated in the presence of exogenous VEGF-A, they form network-like structures in Matrigel and exhibit a cobblestone appearance on collagen IV-coated dishes93. Thus, there are similarities between human ES and iPS cell lines with regard to potential to differentiate towards an endothelial lineage, but whether the regulatory pathways that govern their differentiation are similar remains to be determined. Studies in both human ES and iPS cells, to date, suggest that factors implicated in endothelial cell differentiation are expressed in both cell systems, but in slightly different temporal patterns during the course of differentiation65,67,68,101. Thus, the hierarchy of signaling pathways throughout, and putative intermediate pre-endothelial phenotypes generated during, the differentiation process may differ even though the endothelial cell types ultimately generated from human ES and iPS cells exhibit similar characteristics.

Translation to Clinical Therapy

Cardiovascular disease is the number one cause of death and disability in the US. Understanding the biological activity of stem and progenitor cells, and their ability to contribute to repair, regeneration and remodeling of the heart and blood vessels injured by cardiovascular diseases, is an essential part of the paradigm in enabling us to achieve a reduction in related deaths. In addition, a deeper understanding of the signaling pathways that regulate vascular cell differentiation will enable optimized therapeutic strategies and interventions for many prevalent diseases associated with aberrant blood vessel formation.

Both human ES and iPS cells are promising sources of cells for clinical cardiovascular therapies, as they are able to undergo differentiation towards endothelial cells, vascular smooth muscle cells and cardiomyocytes in vitro102. There are increasing efforts to test these cells in cardiovascular injury models to gain further understanding of their potential and function in vivo. In a recent study tracking the fate of human ES cell-derived endothelial cells, survival of transplanted cells into infarcted murine myocardium was detected at 8 weeks post-infarct, and associated with functional improvement and increased vascular density103. Human ES cell-derived endothelial cells have been shown to integrate into the host circulation69, and enhance revascularization in models of hindlimb ischemia and myocardial infarction103,104. Similarly, iPS cells have been shown to differentiate into cells of the cardiovascular lineages105107, and improve focal cerebral ischemia via subdural transplantation in rats108.

Although these results are promising, continued studies are needed in both in vitro cell culture systems and in vivo translational animal models to gain needed insights into the potential and function of both human ES- and iPS-derived cardiovascular cells, and enable translation from experimental and pre-clinical studies to human clinical therapies.

References

  • 1.Folkman J. Angiogenesis: an organizing principle for drug discovery? Nat Rev Drug Discov. 2007;6:273–286. doi: 10.1038/nrd2115. [DOI] [PubMed] [Google Scholar]
  • 2.Ezekowitz A, Mulliken J, Folkman J. Interferon-α therapy of haemangiomas in newborns and infants. Br J Haematol. 1991;79:67–68. doi: 10.1111/j.1365-2141.1991.tb08123.x. [DOI] [PubMed] [Google Scholar]
  • 3.Szabo S, Folkman J, Vattay P, Morales RE, Pinkus GS, Kato K. Accelerated healing of duodenal ulcers by oral administration of a mutein of basic fibroblast growth factor in rats. Gastroenterology. 1994;106:1106–1111. doi: 10.1016/0016-5085(94)90773-0. [DOI] [PubMed] [Google Scholar]
  • 4.Miller JW, Adamis AP, Shima DT, D’Amore PA, Moulton RS, O’Reilly MS, Folkman J, Dvorak HF, Brown LF, Berse B, Yeo T, Yeo K. Vascular endothelial growth factor/vascular permeability factor is temporally and spatially correlated with ocular angiogenesis in a primate model. Am J Pathol. 1994;145:574–584. [PMC free article] [PubMed] [Google Scholar]
  • 5.Folkman J. In: Targeted Therapies in Rheumatology. Smolen JS, Lipsky PE, editors. Martin Dunitz; London: 2003. pp. 111–131. [Google Scholar]
  • 6.Moulton KS, Vakili K, Zurakowski D, Soliman M, Butterfield C, Sylvin E, Lo KM, Gillies S, Javaherian K, Folkman J. Inhibition of plaque neovascularization reduces macrophage accumulation and progression of advanced atherosclerosis. Proc Natl Acad Sci USA. 2003;100:4736–4741. doi: 10.1073/pnas.0730843100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Moulton KS, Heller E, Konerding MA, Flynn E, Palinski W, Folkman J. Angiogenesis inhibitors endostatin or TNP-470 reduce intimal neovascularization and plaque growth in apolipoprotein E-deficient mice. Circulation. 1999;99:1726–1732. doi: 10.1161/01.cir.99.13.1726. [DOI] [PubMed] [Google Scholar]
  • 8.Moulton KS. Angiogenesis in atherosclerosis: gathering evidence beyond speculation. Curr Opin Lipidol. 2006;17:548–555. doi: 10.1097/01.mol.0000245261.71129.f0. [DOI] [PubMed] [Google Scholar]
  • 9.Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145–1147. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
  • 10.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126:663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 11.Lawson KA, Meneses JJ, Pedersen RA. Clonal analysis of epiblast fate during germ layer formation in the mouse embryo. Development. 1991;113:891–911. doi: 10.1242/dev.113.3.891. [DOI] [PubMed] [Google Scholar]
  • 12.Tam PP, Behringer RR. Mouse gastrulation: the formation of a mammalian body plan. Mech Dev. 1997;68:3–25. doi: 10.1016/s0925-4773(97)00123-8. [DOI] [PubMed] [Google Scholar]
  • 13.Le Lièvre CS, Le Douarin NM. Mesenchymal derivatives of the neural crest: analysis of chimaeric quail and chick embryos. Embryol Exp Morphol. 1975;34:125–154. [PubMed] [Google Scholar]
  • 14.Noden DM. Cell movements and control of patterned tissue assembly during craniofacial development. Craniofac Genet Dev Biol. 1991;11:192–213. [PubMed] [Google Scholar]
  • 15.Pardanaud L, Luton D, Prigent M, Bourcheix LM, Catala M, Dieterlen-Lievre F. Two distinct endothelial lineages in ontogeny, one of them related to hemopoiesis. Development. 1996;122:1363–1371. doi: 10.1242/dev.122.5.1363. [DOI] [PubMed] [Google Scholar]
  • 16.Dyer MA, Farrington SM, Mohn D, Munday JR, Baron MH. Indian hedgehog activates hematopoiesis and vasculogenesis and can respecify prospective neuroectodermal cell fate in the mouse embryo. Development. 2001;128:1717–1730. doi: 10.1242/dev.128.10.1717. [DOI] [PubMed] [Google Scholar]
  • 17.Wurmser AE, Nakashima K, Summers RG, Toni N, D’Amour KA, Lie DC, Gage FH. Cell fusion-independent differentiation of neural stem cells to the endothelial lineage. Nature. 2004;430:350–356. doi: 10.1038/nature02604. [DOI] [PubMed] [Google Scholar]
  • 18.Flamme I, Frölich T, Risau W. Molecular mechanisms of vasculogenesis and embryonic angiogenesis. J Cell Physiol. 1997;173:206–210. doi: 10.1002/(SICI)1097-4652(199711)173:2<206::AID-JCP22>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 19.Patan S. Vasculogenesis and angiogenesis. Cancer Treat Res. 2004;117:3–32. doi: 10.1007/978-1-4419-8871-3_1. [DOI] [PubMed] [Google Scholar]
  • 20.Doetschman TC, Eistetter H, Katz M, Schmidt W, Kemler R. The in vitro development of blastocyst-derived embryonic stem cell lines: formation of visceral yolk sac, blood islands and myocardium. J Embryol Exp Morphol. 1985;87:27–45. [PubMed] [Google Scholar]
  • 21.Becker S, Wang ZJ, Massey H, Arauz A, Labosky P, Hammerschmidt M, St-Jacques B, Bumcrot D, McMahon A, Grabel L. A role for Indian hedgehog in extraembryonic endoderm differentiation in F9 cells and the early mouse embryo. Dev Biol. 1997;187:298–310. doi: 10.1006/dbio.1997.8616. [DOI] [PubMed] [Google Scholar]
  • 22.Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, Fahrig M, Vandenhoeck A, Harpal K, Eberhardt C, Declercq C, Pawling J, Moons L, Collen D, Risau W, Nagy A. Abnormal blood vessel development and lethality in embryos lacking a single VEGF allele. Nature. 1996;380:435–439. doi: 10.1038/380435a0. [DOI] [PubMed] [Google Scholar]
  • 23.Leconte I, Fox JC, Baldwin HS, Buck CA, Swain JL. Adenoviral-mediated expression of antisense RNA to fibroblast growth factors disrupts murine vascular development. Dev Dyn. 1998;213:421–430. doi: 10.1002/(SICI)1097-0177(199812)213:4<421::AID-AJA7>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  • 24.Shing Y, Folkman J, Sullivan R, Butterfield C, Murray J, Klagsbrun M. Heparin affinity: purification of a tumor-derived capillary endothelial cell growth factor. Science. 1984;223:1296–1299. doi: 10.1126/science.6199844. [DOI] [PubMed] [Google Scholar]
  • 25.Shalaby F, Rossant J, Yamaguchi TP, Gertsenstein M, Wu XF, Breitman ML, Schuh AC. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature. 1995;376:62–66. doi: 10.1038/376062a0. [DOI] [PubMed] [Google Scholar]
  • 26.Shalaby F, Ho J, Stanford WL, Fischer KD, Schuh AC, Schwartz L, Bernstein A, Rossant J. A requirement for Flk1 in primitive and definitive hematopoiesis and vasculogenesis. Cell. 1997;89:981–990. doi: 10.1016/s0092-8674(00)80283-4. [DOI] [PubMed] [Google Scholar]
  • 27.Maye P, Becker S, Kasameyer E, Byrd N, Grabel L. Indian hedgehog signaling in extraembryonic endoderm and ectoderm differentiation in ES embryoid bodies. Mech Dev. 2000;94:117–132. doi: 10.1016/s0925-4773(00)00304-x. [DOI] [PubMed] [Google Scholar]
  • 28.Winnier G, Blessing M, Labosky PA, Hogan BL. Bone morphogenetic protein-4 is required for mesoderm formation and patterning in the mouse. Genes Dev. 1995;9:2105–2116. doi: 10.1101/gad.9.17.2105. [DOI] [PubMed] [Google Scholar]
  • 29.Hollenhorst PC, Jones DA, Graves BJ. Expression profiles frame the promoter specificity dilemma of the ETS family of transcription factors. Nucleic Acids Res. 2004;32:5693–5702. doi: 10.1093/nar/gkh906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Bernat JA, Crawford GE, Ogurtsov AY, Collins FS, Ginsburg D, Kondrashov AS. Distant conserved sequences flanking endothelial-specific promoters contain tissue-specific Dnase-hypersensitive sites and over-represented motifs. Hum Mol Genet. 2006;15:2098–2105. doi: 10.1093/hmg/ddl133. [DOI] [PubMed] [Google Scholar]
  • 31.De Val S, Chi NC, Meadows SM, Minovitsky S, Anderson JP, Harris IS, Ehlers ML, Agarwal P, Visel A, Xu SM, Pennacchio LA, Dubchak I, Krieg PA, Stainier DY, Black BL. Combinatorial regulation of endothelial gene expression by Ets and Forkhead transcription factors. Cell. 2008;135:1053–1064. doi: 10.1016/j.cell.2008.10.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Barton K, Muthusamy N, Fischer C, Ting CN, Walunas TL, Lanier LL, Leiden JM. The Ets-1 transcription factor is required for the development of natural killer cells in mice. Immunity. 1998;9:555–563. doi: 10.1016/s1074-7613(00)80638-x. [DOI] [PubMed] [Google Scholar]
  • 33.Hart A, Melet F, Grossfeld P, Chien K, Jones C, Tunnacliffe A, Favier R, Bernstein A. Fli-1 is required for murine vascular and megakaryocytic development and is hemizygously deleted in patients with thrombocytopenia. Immunity. 2000;13:167–177. doi: 10.1016/s1074-7613(00)00017-0. [DOI] [PubMed] [Google Scholar]
  • 34.Pham VN, Lawson ND, Mugford JW, Dye L, Castranova D, Lo B, Weinstein BM. Combinatorial function of ETS transcription factors in the developing vasculature. Dev Biol. 2007;303:772–783. doi: 10.1016/j.ydbio.2006.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Spyropoulos DD, Pharr PN, Lavenburg KR, Jackers P, Papas TS, Ogawa M, Watson DK. Hemorrhage, impaired hematopoiesis, and lethality in mouse embryos carrying a targeted disruption of the Fli1 transcription factor. Mol Cell Biol. 2000;20:5643–5652. doi: 10.1128/mcb.20.15.5643-5652.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wang LC, Kuo F, Fujiwara Y, Gilliland DG, Golub TR, Orkin SH. Yolk sac angiogenic defect and intra-embryonic apoptosis in mice lacking the Ets-related factor TEL. EMBO J. 1997;16:4374–4383. doi: 10.1093/emboj/16.14.4374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ferdous A, Caprioli A, Iacovino M, Martin CM, Morris J, Richardson JA, Latif S, Hammer RE, Harvey RP, Olson EN, Kyba M, Garry DJ. Nkx2-5 transactivates the Ets-related protein 71 gene and specifies an endothelial/endocardial fate in the developing embryo. Proc Natl Acad Sci USA. 2009;106:814–819. doi: 10.1073/pnas.0807583106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lee D, Park C, Lee H, Lugus JJ, Kim SH, Arentson E, Chung YS, Gomez G, Kyba M, Lin S, Janknecht R, Lim DS, Choi K. ER71 acts downstream of BMP, Notch, and Wnt signaling in blood and vessel progenitor specification. Cell Stem Cell. 2008;2:497–507. doi: 10.1016/j.stem.2008.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Sumanas S, Gomez G, Zhao Y, Park C, Choi K, Lin S. Interplay among Etsrp/ER71, Scl, and Alk8 signaling controls endothelial and myeloid cell formation. Blood. 2008;111:4500–4510. doi: 10.1182/blood-2007-09-110569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Salanga MC, Meadows SM, Myers CT, Krieg PA. ETS family protein ETV2 is required for initiation of the endothelial lineage but not the hematopoietic lineage in the Xenopus embryo. Dev Dyn. 2010;239:1178–1187. doi: 10.1002/dvdy.22277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Furuyama T, Kitayama K, Shimoda Y, Ogawa M, Sone K, Yoshida-Araki K, Hisatsune H, Nishikawa S, Nakayama K, Nakayama K, Ikeda K, Motoyama N, Mori N. Abnormal angiogenesis in Foxo1 (Fkhr)-deficient mice. J Biol Chem. 2004;279:34741–34749. doi: 10.1074/jbc.M314214200. [DOI] [PubMed] [Google Scholar]
  • 42.Hosaka T, Biggs WH, 3rd, Tieu D, Boyer AD, Varki NM, Cavenee WK, Arden KC. Disruption of forkhead transcription factor (FOXO) family members in mice reveals their functional diversification. Proc Natl Acad Sci USA. 2004;101:2975–2980. doi: 10.1073/pnas.0400093101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Mahlapuu M, Ormestad M, Enerback S, Carlsson P. The forkhead transcription factor Foxf1 is required for differentiation of extra-embryonic and lateral plate mesoderm. Development. 2001;128:155–166. doi: 10.1242/dev.128.2.155. [DOI] [PubMed] [Google Scholar]
  • 44.Kume T, Jiang H, Topczewska JM, Hogan BL. The murine winged helix transcription factors, Foxc1 and Foxc2, are both required for cardiovascular development and somitogenesis. Genes Dev. 2001;15:2470–2482. doi: 10.1101/gad.907301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Astorga J, Carlsson P. Hedgehog induction of murine vasculogenesis is mediated by Foxf1 and Bmp4. Development. 2007;134:3753–3761. doi: 10.1242/dev.004432. [DOI] [PubMed] [Google Scholar]
  • 46.Hayashi H, Kume T. Foxc transcription factors directly regulate Dll4 and Hey2 expression by interacting with the VEGF-Notch signaling pathways in endothelial cells. PLoS ONE. 2008;3:e2401. doi: 10.1371/journal.pone.0002401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Seo S, Fujita H, Nakano A, Kang M, Duarte A, Kume T. The forkhead transcription factors, Foxc1 and Foxc2, are required for arterial specification and lymphatic sprouting during vascular development. Dev Biol. 2006;294:458–470. doi: 10.1016/j.ydbio.2006.03.035. [DOI] [PubMed] [Google Scholar]
  • 48.Atkins GB, Jain MK. Role of Kruppel-like transcription factors in endothelial biology. Circ Res. 2007;100:1686–1695. doi: 10.1161/01.RES.0000267856.00713.0a. [DOI] [PubMed] [Google Scholar]
  • 49.Tsai S, Martin SI, Zon LI, D’Andrea AD, Wong GG, Orkin SH. Cloning of cDNA for the major DNA-binding protein of the erythroid lineage through expression in mammalian cells. Nature. 1989;339:446–451. doi: 10.1038/339446a0. [DOI] [PubMed] [Google Scholar]
  • 50.Lugus JJ, Chung YS, Mills JC, Kim SI, Grass J, Kyba M, Doherty JM, Bresnick EH, Choi K. GATA2 functions at multiple steps in hemangioblast development and differentiation. Development. 2007;134:393–405. doi: 10.1242/dev.02731. [DOI] [PubMed] [Google Scholar]
  • 51.Geriling M, Rodaway ARF, Gottgens B, Patient RK, Green AR. The SCL gene specifies haemangioblast development from early mesoderm. EMBO J. 1998;17:4029–4045. doi: 10.1093/emboj/17.14.4029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Mead P, Kelley CM, Hahn PS, Piedad O, Zon LI. SCL specifies hematopoietic mesoderm in Xenopus embryos. Development. 1998;125:2611–2620. doi: 10.1242/dev.125.14.2611. [DOI] [PubMed] [Google Scholar]
  • 53.Deleuze V, Chalhoub E, El-Hajj R, Dohet C, Le Clech M, Couraud PO, Huber P, Mathieu D. TAL-1/SCL and its partners E47 and LMO2 up-regulate VE-cadherin expression in endothelial cells. Mol Cell Biol. 2007;27:2687–2697. doi: 10.1128/MCB.00493-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Xiong J, Leahy A, Lee HH, Stuhlmann H. Vezf1: A Zn finger transcription factor restricted to endothelial cells and their precursors. Dev Biol. 1999;206:123–141. doi: 10.1006/dbio.1998.9144. [DOI] [PubMed] [Google Scholar]
  • 55.Kuhnert F, Campagnolo L, Xiong JW, Lemons D, Fitch MJ, Zou Z, Kiosses WB, Gardner H, Stuhlmann H. Dosage-dependent requirement for mouse Vezf1 in vascular system development. Dev Biol. 2005;283:140–156. doi: 10.1016/j.ydbio.2005.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hansis C. Totipotency, cell differentiation and reprogramming in humans. Reprod Biomed Online. 2006;13:551–557. doi: 10.1016/s1472-6483(10)60644-x. [DOI] [PubMed] [Google Scholar]
  • 57.Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R, Slukvin II, Thomson JA. Induced pluripotent stem cell lines derived from human somatic cells. Science. 2007;318:1917–1920. doi: 10.1126/science.1151526. [DOI] [PubMed] [Google Scholar]
  • 58.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131:861–872. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 59.Nistor GI, Totoiu MO, Haque N, Carpenter MK, Keirstead HS. Human embryonic stem cells differentiate into oligodendrocytes in high purity and myelinate after spinal cord transplantation. Glia. 2005;49:385–396. doi: 10.1002/glia.20127. [DOI] [PubMed] [Google Scholar]
  • 60.Freund C, Ward-van Oostwaard D, Monshouwer-Kloots J, van den Brink S, van Rooijen M, Xu X, Zweigerdt R, Mummery C, Passier R. Insulin redirects differentiation from cardiogenic mesoderm and endoderm to neuroectoderm in differentiating human embryonic stem cells. Stem Cells. 2008;26:724–733. doi: 10.1634/stemcells.2007-0617. [DOI] [PubMed] [Google Scholar]
  • 61.Laflamme MA, Gold J, Xu C, Hassanipour M, Rosler E, Police S, Muskheli V, Murry CE. Formation of human myocardium in the rat heart from human embryonic stem cells. Am J Pathol. 2005;167:663–671. doi: 10.1016/S0002-9440(10)62041-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Saeki K, Saeki K, Nakahara M, Matsuyama S, Nakamura N, Yogiashi Y, Yoneda A, Koyanagi M, Kondo Y, Yuo A. A feeder-free and efficient production of functional neutrophils from human embryonic stem cells. Stem Cells. 2009;27:59–67. doi: 10.1634/stemcells.2007-0980. [DOI] [PubMed] [Google Scholar]
  • 63.Lavon N, Yanuka O, Benvenisty N. Differentiation and isolation of hepatic-like cells from human embryonic stem cells. Differentiation. 2004;72:230–238. doi: 10.1111/j.1432-0436.2004.07205002.x. [DOI] [PubMed] [Google Scholar]
  • 64.Lavon N, Yanuka O, Benvenisty N. The effect of overexpression of Pdx1 and Foxa2 on the differentiation of human embryonic stem cells into pancreatic cells. Stem Cells. 2006;24:1923–1930. doi: 10.1634/stemcells.2005-0397. [DOI] [PubMed] [Google Scholar]
  • 65.Levenberg S, Golub JS, Amit M, Itskovitz-Eldor J, Langer R. Endothelial cells derived from human embryonic stem cells. Proc Natl Acad Sci USA. 2002;99:4391–4396. doi: 10.1073/pnas.032074999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wang L, Li L, Shojaei F, Levac K, Cerdan C, Menendez P, Martin T, Rouleau A, Bhatia M. Endothelial and hematopoietic cell fate of human embryonic stem cells originates from primitive endothelium with hemangioblastic properties. Immunity. 2004;21:31–41. doi: 10.1016/j.immuni.2004.06.006. [DOI] [PubMed] [Google Scholar]
  • 67.Vodyanik MA, Bork JA, Thomson JA, Slukvin II. Human embryonic stem cell-derived CD34+ cells: efficient production in the coculture with OP9 stromal cells and analysis of lymphohematopoietic potential. Blood. 2005;105:617–626. doi: 10.1182/blood-2004-04-1649. [DOI] [PubMed] [Google Scholar]
  • 68.Zambidis ET, Peault B, Park TS, Bunz F, Civin CI. Hematopoietic differentiation of human embryonic stem cells progresses through sequential hematoendothelial, primitive, and definitive stages resembling human yolk sac development. Blood. 2005;106:860–870. doi: 10.1182/blood-2004-11-4522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lu SJ, Feng Q, Caballero S, Chen Y, Moore MA, Grant MB, Lanza R. Generation of functional hemangioblasts from human embryonic stem cells. Nat Methods. 2007;4:501–509. doi: 10.1038/nmeth1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Baldwin H, Shen HM, Yan H-C, DeLisser HM, Chung AM, Trask T, Kirshbaum NE, Newman PJ, Albelda SM, Buck CA. Platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31): alternatively spliced, functionally distinct isoforms expressed during mammalian cardiovascular development. Development. 1994;120:2539–2553. doi: 10.1242/dev.120.9.2539. [DOI] [PubMed] [Google Scholar]
  • 71.DeLisser H, Newman PJ, Albelda SM. Molecular and functional aspects of PECAM-1/CD31. Immunology Today. 1994;15:490–495. doi: 10.1016/0167-5699(94)90195-3. [DOI] [PubMed] [Google Scholar]
  • 72.Pinter E, Barreuther M, Lu T, Imhof BA, Madri JA. Platelet-endothelial cell adhesion molecule-1(PECAM-1/CD31) tyrosine phosphorylation state changes during vasculogenesis in the murine conceptus. Am J Pathol. 1997;150:1523–1530. [PMC free article] [PubMed] [Google Scholar]
  • 73.Breier G, Breviario F, Caveda L, Berthier R, Schnurch H, Gotsch U, Vestweber D, Risau W, Dejana E. Molecular cloning and expression of murine vascular endothelial-cadherin in early stage development of cardiovascular system. Blood. 1996;87:630–641. [PubMed] [Google Scholar]
  • 74.Pera MF, Trounson AO. Human embryonic stem cells: prospects for development. Development. 2004;131:5515–5525. doi: 10.1242/dev.01451. [DOI] [PubMed] [Google Scholar]
  • 75.Chadwick K, Wang L, Li L, Menendez P, Murdoch B, Rouleau A, Bhatia M. Cytokines and BMP-4 promote hematopoietic differentiation of human embryonic stem cells. Blood. 2003;102:906–915. doi: 10.1182/blood-2003-03-0832. [DOI] [PubMed] [Google Scholar]
  • 76.Nakano T, Kodama H, Honjo T. Generation of lymphohematopoietic cells from embryonic stem cells in culture. Science. 1994;265:1098–1101. doi: 10.1126/science.8066449. [DOI] [PubMed] [Google Scholar]
  • 77.Kelly MA, Hirschi KK. Signaling Hierarchy Regulating Human Endothelial Cell Development. Arterioscler Thromb Vasc Biol. 2009;29:718–724. doi: 10.1161/ATVBAHA.109.184200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Xu C, Inokuma MS, Denham J, Golds K, Kundu P, Gold JD, Carpenter MK. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol. 2001;19:971–974. doi: 10.1038/nbt1001-971. [DOI] [PubMed] [Google Scholar]
  • 79.Ludwig TE, Bergendahl V, Levenstein ME, Yu J, Probasco MD, Thomson JA. Feeder-independent culture of human embryonic stem cells. Nat Methods. 2006;3:637–646. doi: 10.1038/nmeth902. [DOI] [PubMed] [Google Scholar]
  • 80.Unger C, Gao S, Cohen M, Jaconi M, Bergstrom R, Holm F, Galan A, Sanchez E, Irion O, Dubuisson JB, Giry-Laterriere M, Salmon P, Simon C, Hovatta O, Feki A. Immortalized human skin fibroblast feeder cells support growth and maintenance of both human embryonic and induced pluripotent stem cells. Hum Reprod. 2009;24:2567–2581. doi: 10.1093/humrep/dep232. [DOI] [PubMed] [Google Scholar]
  • 81.Sieveking DP, Ng MK. Cell therapies for therapeutic angiogenesis: back to the bench. Vasc Med. 2009;14:153–166. doi: 10.1177/1358863X08098698. [DOI] [PubMed] [Google Scholar]
  • 82.Nakagawa M, Koyanagi M, Tanabe K, Takahashi K, Ichisaka T, Aoi T, Okita K, Mochiduki Y, Takizawa N, Yamanaka S. Generation of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol. 2008;26:101–106. doi: 10.1038/nbt1374. [DOI] [PubMed] [Google Scholar]
  • 83.Okita K, Ichisaka T, Yamanaka S. Generation of germline-competent induced pluripotent stem cells. Nature. 2007;448:313–317. doi: 10.1038/nature05934. [DOI] [PubMed] [Google Scholar]
  • 84.Wernig M, Lengner CJ, Hanna J, Lodato MA, Steine E, Foreman R, Staerk J, Markoulaki S, Jaenisch R. A drug-inducible transgenic system for direct reprogramming of multiple somatic cell types. Nat Biotechnol. 2008;26:916–924. doi: 10.1038/nbt1483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S. Generation of Mouse Induced Pluripotent Stem Cells Without Viral Vectors. Science. 2008;322:949–953. doi: 10.1126/science.1164270. [DOI] [PubMed] [Google Scholar]
  • 86.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;458:766–770. doi: 10.1038/nature07864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Woltjen K, Michael IP, Mohseni P, Desai R, Mileikovsky M, Hämäläinen R, Cowling R, Wang W, Liu P, Gertsenstein M, Kaji K, Sung HK, Nagy A. piggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature. 2009;458:771–775. doi: 10.1038/nature07863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Soldner F, Hockemeyer D, Beard C, Gao Q, Bell GW, Cook EG, Hargus G, Blak A, Cooper O, Mitalipova M, Isacson O, Jaenisch R. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell. 2009;136:964–977. doi: 10.1016/j.cell.2009.02.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Chang CW, Lai YS, Pawlik KM, Liu K, Sun CW, Li C, Schoeb TR, Townes TM. Polycistronic lentiviral vector for “hit and run” reprogramming of adult skin fibroblasts to induced pluripotent stem cells. Stem Cells. 2009;27:1042–1049. doi: 10.1002/stem.39. [DOI] [PubMed] [Google Scholar]
  • 90.Yu J, Hu K, Smuga-Otto K, Tian S, Stewart R, Slukvin II, Thomson JA. Human induced pluripotent stem cells free of vector and transgene sequences. Science. 2009;324:797–801. doi: 10.1126/science.1172482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Zhou H, Wu S, Joo JY, Zhu S, Han DW, Lin T, Trauger S, Bien G, Yao S, Zhu Y, Siuzdak G, Schöler HR, Duan L, Ding S. Generation of Induced Pluripotent Stem Cells Using Recombinant Proteins. Cell Stem Cell. 2009;4:381–384. doi: 10.1016/j.stem.2009.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zhou W, Freed CR. Adenoviral Gene Delivery Can Reprogram Human Fibroblasts to Induced Pluripotent Stem Cells. Stem Cells. 2009;27:2667–2674. doi: 10.1002/stem.201. [DOI] [PubMed] [Google Scholar]
  • 93.Choi KD, Yu J, Smuga-Otto K, Salvagiotto G, Rehrauer W, Vodyanik M, Thomson J, Slukvin I. Hematopoietic and endothelial differentiation of human induced pluripotent stem cells. Stem Cells. 2009;27:559–567. doi: 10.1634/stemcells.2008-0922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Taura D, Sone M, Homma K, Oyamada N, Takahashi K, Tamura N, Yamanaka S, Nakao K. Induction and isolation of vascular cells from human induced pluripotent stem cells-brief report. Arterioscler Thromb Vasc Biol. 2009;29:1100–1103. doi: 10.1161/ATVBAHA.108.182162. [DOI] [PubMed] [Google Scholar]
  • 95.Poon E, Clermont F, Firpo MT, Akhurst RJ. TGFbeta inhibition of yolk-sac-like differentiation of human embryonic stem-cell-derived embryoid bodies illustrates differences between early mouse and human development. J Cell Sci. 2006;119:759–768. doi: 10.1242/jcs.02788. [DOI] [PubMed] [Google Scholar]
  • 96.Ginis I, Luo Y, Miura T, Thies S, Brandenberger R, Gerecht-Nir S, Amit M, Hoke A, Carpenter MK, Itskovitz-Eldor J, Rao MS. Differences between human and mouse embryonic stem cells. Dev Biol. 2004;269:360–380. doi: 10.1016/j.ydbio.2003.12.034. [DOI] [PubMed] [Google Scholar]
  • 97.Baron MH. Molecular regulation of embryonic hematopoiesis and vascular development: a novel pathway. J Hematother Stem Cell Res. 2001;10:587–594. doi: 10.1089/152581601753193797. [DOI] [PubMed] [Google Scholar]
  • 98.Byrd N, Becker S, Maye P, Narasimhaiah R, St-Jacques B, Zhang X, McMahon J, McMahon A, Grabel L. Hedgehog is required for murine yolk sac angiogenesis. Development. 2002;129:361–372. doi: 10.1242/dev.129.2.361. [DOI] [PubMed] [Google Scholar]
  • 99.St-Jacques B, Hammerschmidt M, McMahon AP. Indian hedgehog signaling regulates proliferation and differentiation of chondrocytes and is essential for bone formation. Genes Dev. 1999;13:2072–2086. doi: 10.1101/gad.13.16.2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Oberlin E, Tavian M, Blazsek I, Péault B. Blood-forming potential of vascular endothelium in the human embryo. Development. 2002;129:4147–4157. doi: 10.1242/dev.129.17.4147. [DOI] [PubMed] [Google Scholar]
  • 101.Schenke-Layland K, Rhodes KE, Angelis E, Butylkova Y, Heydarkhan-Hagvall S, Gekas C, Zhang R, Goldhaber JI, Mikkola HK, Plath K, MacLellan WR. Reprogrammed mouse fibroblasts differentiate into cells of the cardiovascular and hematopoietic lineages. Stem Cells. 2008;26:1537–1546. doi: 10.1634/stemcells.2008-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Nazaraki G, Uosaki H, Teranishi M, Okita K, Kim B, Matsuoka S, Yamanaka S, Yamashita JK. Directed and systematic differentiation of cardiovascular cells from mouse induced pluripotent stem cells. Circulation. 2008;118:498–506. doi: 10.1161/CIRCULATIONAHA.108.769562. [DOI] [PubMed] [Google Scholar]
  • 103.Li Z, Wilson KD, Smith B, Kraft DL, Jia F, Huang M, Xie X, Robbins RC, Gambhir SS, Weissman IL, Wu JC. Differentiation, survival, and function of embryonic stem cell derived endothelial cells for ischemic heart disease. PLoS One. 2009;4:e8443. doi: 10.1371/journal.pone.0008443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Sone M, Itoh H, Yamahara K, Yamashita JK, Yurugi-Kobayashi T, Nonoguchi A, Suzuki Y, Chao TH, Sawada N, Fukunaga Y, Miyashita K, Park K, Oyamada N, Sawada N, Taura D, Tamura N, Kondo Y, Nito S, Suemori H, Nakatsuji N, Nishikawa S, Nakao K. Pathway for differentiation of human embryonic stem cells to vascular cell components and their potential for vascular regeneration. Arterioscler Thromb Vasc Biol. 2007;27:2127–2134. doi: 10.1161/ATVBAHA.107.143149. [DOI] [PubMed] [Google Scholar]
  • 105.Germanguz I, Sedan O, Zeevi-Levin N, Shtreichman R, Barak E, Ziskind A, Eliyahu S, Meiry G, Amit M, Itskovitz-Eldor J, Binah O. Molecular characterization and functional properties of cardiomyocytes derived from human inducible pluripotent stem cells. J Cell Mol Med. 2009 Dec 11; doi: 10.1111/j.1582-4934.2009.00996.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Mauritz C, Schwanke K, Reppel M, Neef S, Katsirntaki K, Maier LS, Nguemo F, Menke S, Haustein M, Hescheler J, Hasenfuss G, Martin U. Generation of functional murine cardiac myocytes from induced pluripotent stem cells. Circulation. 2008;118:507–517. doi: 10.1161/CIRCULATIONAHA.108.778795. [DOI] [PubMed] [Google Scholar]
  • 107.Zhang J, Wilson GF, Soerens AG, Koonce CH, Yu J, Palecek SP, Thomson JA, Kamp TJ. Functional cardiomyocytes derived from human induced pluripotent stem cells. Circ Res. 2009;104:e30–41. doi: 10.1161/CIRCRESAHA.108.192237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Chen SJ, Chang CM, Tsai SK, Chang YL, Chou SJ, Huang SS, Tai LK, Chen YC, Ku HH, Li HY, Chiou SH. Functional Improvement of Focal Cerebral Ischemia Injury by Subdural Transplantation of Induced Pluripotent Stem Cells with Fibrin Glue. Stem Cells Dev. 2010 Mar 1; doi: 10.1089/scd.2009.0452. [DOI] [PubMed] [Google Scholar]
  • 109.Farrington SM, Belaoussoff M, Baron MH. Winged-helix, Hedgehog and BMP genes are differentially expressed in distinct cell layers of the murine yolk sac. Mech Dev. 1997;62:197–211. doi: 10.1016/s0925-4773(97)00664-3. [DOI] [PubMed] [Google Scholar]
  • 110.Grabel L, Becker S, Lock L, Maye P, Zanders T. Using EC and ES cell culture to study early development: recent observations on Indian hedgehog and BMPs. Int J Dev Biol. 1998;42:917–925. [PubMed] [Google Scholar]
  • 111.Johansson BM, Wiles MV. Evidence for involvement of activin A and bone morphogenetic protein 4 in mammalian mesoderm and hematopoietic development. Mol Cell Biol. 1995;15:141–151. doi: 10.1128/mcb.15.1.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Zhang P, Li J, Tan Z, Wang C, Liu T, Chen L, Yong J, Jiang W, Sun X, Du L, Ding M, Deng H. Short-term BMP-4 treatment initiates mesoderm induction in human embryonic stem cells. Blood. 2008;111:1933–1941. doi: 10.1182/blood-2007-02-074120. [DOI] [PubMed] [Google Scholar]
  • 113.Pick M, Azzola L, Mossman A, Stanley EG, Elefanty AG. Differentiation of human embryonic stem cells in serum-free medium reveals distinct roles for bone morphogenetic protein 4, vascular endothelial growth factor, stem cell factor, and fibroblast growth factor 2 in hematopoiesis. Stem Cells. 2007;25:2206–2214. doi: 10.1634/stemcells.2006-0713. [DOI] [PubMed] [Google Scholar]
  • 114.Yamaguchi TP, Conlon RA, Rossant J. Expression of the fibroblast growth factor receptor FGFR-1/flg during gastrulation and segmentation in the mouse embryo. Dev Biol. 1992;152:75–88. doi: 10.1016/0012-1606(92)90157-c. [DOI] [PubMed] [Google Scholar]
  • 115.Miquerol L, Gertsenstein M, Harpal K, Rossant J, Nagy A. Multiple developmental roles of VEGF suggested by a LacZ-tagged allele. Dev Biol. 1999;212:307–322. doi: 10.1006/dbio.1999.9355. [DOI] [PubMed] [Google Scholar]
  • 116.Anderson KP, Kern CB, Crable SC, Lingrel JB. Isolation of a gene encoding a functional zinc finger protein homologous to erythroid Kruppel-like factor: identification of a new multigene family. Mol Cell Biol. 1995;15:5957–5965. doi: 10.1128/mcb.15.11.5957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Gomez GA, Veldman MB, Zhao Y, Burgess S, Lin S. Discovery and characterization of novel vascular and hematopoietic genes downstream of etsrp in zebrafish. PLoS One. 2009;4:e4994. doi: 10.1371/journal.pone.0004994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Kallianpur AR, Jordan JE, Brandt SJ. The SCL/TAL-1 gene is expressed in progenitors of both the hematopoietic and vascular systems during embryogenesis. Blood. 1994;83:1200–1208. [PubMed] [Google Scholar]

RESOURCES