Abstract
Ischemic cardiovascular disease still remains as a leading cause of morbidity and mortality despite various medical, surgical, and interventional therapy. As such, cell therapy has emerged as an attractive option because it tackles underlying problem of the diseases by inducing neovascularization in ischemic tissue. After overall failure of adult stem or progenitor cells, studies attempted to generate endothelial cells (ECs) from pluripotent stem cells (PSCs). While endothelial cells (ECs) differentiated from PSCs successfully induced vascular regeneration, differentiating volatility and tumorigenic potential is a concern for their clinical applications. Alternatively, direct reprogramming strategies employ lineage-specific factors to change cell fate without achieving pluripotency. ECs have been successfully reprogrammed via ectopic expression of transcription factors (TFs) from endothelial lineage. The reprogrammed ECs induced neovascularization in vitro and in vivo and thus demonstrated their therapeutic value in animal models of vascular insufficiency. Methods of delivering reprogramming factors include lentiviral or retroviral vectors and more clinically relevant, non-integrative adenoviral and episomal vectors. Most studies made use of fibroblast as a source cell for reprogramming, but reprogrammability of other clinically relevant source cell types has to be evaluated. Specific mechanisms and small molecules that are involved in the aforementioned processes tackles challenges associated with direct reprogramming efficiency and maintenance of reprogrammed EC characteristics. After all, this review provides summary of past and contemporary methods of direct endothelial reprogramming and discusses the future direction to overcome these challenges to acquire clinically applicable reprogrammed ECs.
Keywords: cardiovascular disease, Neovascularization, Endothelial cells, Direct reprogramming, Regenerative medicine, Cell Therapy
1. Introduction
Ischemic cardiovascular diseases are the leading causes of morbidity and mortality [1]. Treatment options for chronic, severe cases of ischemia such as myocardial infarction and critical limb ischemia are still limited. Despite significant improvement in surgical, interventional, and medical therapeutics, patients with advanced cases have poor prognoses even after exhaustive treatment with conventional therapeutics leaving them no other option but cardiac transplantation or lower extremity amputation [2–8]. Therefore, novel approaches have been demanded for restoring proper blood perfusion and tissue repair.
Underlying burden of the coronary and peripheral artery diseases are loss of blood vessels and inability to restore vessels with endogenous mechanisms. Cell therapy has been considered an attractive strategy as it supplies vascular components, especially endothelial cells (ECs), for generation of new functional blood vessels (neovascularization). Early attempts of cell therapy was done with various types of adult stem or progenitor cells. Mesenchymal stem cells, mononuclear cells or endothelial progenitor cells derived from bone marrow or peripheral blood have shown to secrete proangiogenic and cytoprotective factors but are modestly effective, lacking evidence of major transdifferentiation of these cell types into ECs [9–12]. Therefore, researchers have sought to generate ECs which can induce neovascularization with different cell sources.
Pluripotent stem cells (PSCs) including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) emerged as a method of generating ECs. Particularly, iPSCs have been considered as a more practical candidate over ESCs as they are free from ethical concerns and can be generated from autologous sources. Ever since Yamanaka and colleagues first reported generation of iPSCs with four transcription factors (TFs), differentiation techniques for generating ECs have significantly improved [13–16]. Multiple studies showed differentiation of human iPSCs (hiPSCs) into ECs and these hiPSC-ECs demonstrated excellent therapeutic potential and neovascularization capacity in ischemic murine models [17, 18]. However, complicated differentiation steps and difficulties in phenotype maintenance have delayed clinical translation of iPSC-derived cells [19]. More importantly, residual, undifferentiated or unwantedly differentiated iPSCs which may exist within the differentiated cell cultures may have tumorigenic potential and aberrant tissue formation [20].
To avoid issues associated with PSCs, novel methods of generating target cell types using lineage-specific TFs or small molecules have been developed. These methods directly convert various source cells to target cell types while bypassing the pluripotent states [21]. This novel approach referred to as “direct reprogramming” or “transdifferentiation” provides significant advantages as it provides time- and cost-effective ways of generating target cells while reducing potential risks and inefficiency associated with pluripotent stem cells.
In this review, we dissect methods of directly reprogramming ECs and characteristics of the reprogrammed or induced ECs. We further discuss therapeutic potential, challenges, and future directions of EC reprogramming.
2. Pathway to direct endothelial reprogramming
2.1. Partial activation of pluripotency
To overcome the problems of PSCs, alternative methods of generating ECs while avoiding or being minimally affected by the pluripotent state have been developed. In 2012, Margariti et al. transiently induced the expression of iPSC-inducing factors OCT4, SOX2, KLF4, and c-MYC (OSKM) to convert human fibroblasts into a dedifferentiated state referred to as partial-iPSCs (PiPSCs) [22]. Four-days of induction of OSKM was sufficient to alter plasticity of the fibroblasts, and subsequent cell culture under endothelial cell growth media-2 (EGM-2) differentiated PiPSCs into ECs (PiPSC-ECs). Unlike iPSCs, tumors were undetected 2 months after PiPSCs were injected into mice with Matrigel. Similarly, Kurian et al. generated a plastic intermediate state from human fetal and adult fibroblasts by introducing miR302 and miR307 with either 4 factors (OSKM) or 6 factors (OCT4, SOX2, KLF4, LMYC, LIN28, p53 short hairpin RNA) for 8 days [23]. These plastic cells were cultured under mesodermal induction medium for 8 days to generate CD34+ angioblast-like progenitor cells. Another 8-day culture under EGM-2 culture medium differentiated the progenitor cells into ECs. Notably, pluripotency markers TRA-1–60 and TRA-1–81 were undetectable even after plastic state induction.
Interestingly, Li et al. only used two pluripotent TFs, OCT4 and KLF4, and chemically defined media to generate induced ECs (iECs) [24]. After lentiviral transduction of these two TFs, the cells were cultured with bone morphogenetic protein 4 (BMP4) for first 7 days, cAMP-dependent protein kinase A (8-Br-cAMP) for the next 7 days, and TGF-beta inhibitor (SB43152) for another 14 days. They noted the pluripotency network was minimally affected by the gradual transdifferentiation steps as suggested from the analysis of transcriptomic and epigenetic changes. Taken together, partial or transient activation of pluripotency reduces risks of tumorigenesis as it bypasses the fully pluripotent state while expediting reprogramming procedure for EC generation. However, achieving dedifferentiated or a partial pluripotent state still requires overexpression of pluripotency reprogramming factors, and therefore, is not completely free from the risks posed by PSCs.
2.2. Direct endothelial reprogramming
Inducing cellular reprogramming toward PSCs by overexpression of pluripotency TFs hinted the possibility of direct cellular reprogramming or fate conversion toward ECs by lineage-specific factors. Thus far, ~15 studies were published in endothelial cell generation via direct reprogramming, and we summarized them in Figure 1 and Table 1.
Figure 1.
Methods of direct endothelial reprogramming with various source cell types. Fibroblasts, human adipose-derived stem cells (hADSCs), mesenchymal stem cells (hMSCs), and human amniotic fluid-derived cells (hACs) were reprogrammed into endothelial cells (ECs). A. The diagram showing origin of the source cells. B. The graphic showing source cell types, reprogramming methods, and target cell. Alphabets from A-N indicate studies as referred from Table 1.
Table 1.
Summary of direct endothelial reprogramming methods
# | Reprogrammed cell name | Source cell type | Key factors | Delivery method | Culture condition | Culture duration | Reprogramming efficiency | In vivo animal model (follow up) | Reference | |
---|---|---|---|---|---|---|---|---|---|---|
Genes | Small molecules | |||||||||
A | rAC-VEC | Human amniotic fluid-derived cell | ETV2, FLI1, and ERG1 | VEGF and SB431542 | Lentiviral infection | EM: SB431542, EC supplement, and Heparin | 28 days | D14: 39.1% CDH5+, 8.0% KDR+ D21: 86.5% CDH5+, 48.5% PECAM1+ | Matrigel Plug (2 weeks) Liver engraftment assay (3 months) | [26] |
B | iEC | Mouse skin and tail-tip fibroblast | Etv2, Foxo1, Klf2, Tal1 and Lmo2 | Lentiviral infection | EBM-2 | 12 days | D12: 4% Tie2-GFP+, 2.3% PECAM1+, 4.2% CDH5+, 2.6% KDR+, 7.3% vWF+ | HLI model LDPI & IHC (2 weeks) | [25] | |
C | iEC* | Human neonatal dermal fibroblast | (Poly I:C) bFGF, VEGF, BMP4, 8-Br- cAMP, SB43152 | Chemical stimulation |
Activation of innate immunity: Poly I:C Transdifferentiation medium I: bFGF, VEGF, and BMP4 Transdifferentiation medium II: EGM-2, bFGF, VEGF, BMP4, and 8-Br-cAMP After sorting: EGM-2 and SB431542 |
7 days + 7 days + 14 days and more | D14: 2% PECAM1+ | Matrigel Plug (2 Weeks) HLI Model 1 st injection immediately after HLI surgery 2nd injection 10 days after HLI surgery LDPI (14 days) & IHC (18 days) | [28] | |
D | ETVEC | Human adult fibroblast | ETV2 | VEGF and bFGF | Lentiviral infection | EGM-2, VEGF, and bFGF | 25 days (beyond 50 days) | D14: 40% KDR+ D15: 3.4% CDH5+ & PECAM1+ D25: 95.3% PECAM1+ |
Matrigel Plug (2 weeks and 6 weeks) HLI model (2 weeks) |
[27] |
E | iEC | Human neonatal fibroblast | ETV2, FLI1, GATA2, and KLF4 | BMP4, VEGF, bFGF, and SB431542 | Lentiviral infection |
Differentiation medium: BMP4, VEGF, and bFGF EC growth medium: EGM-2 MV and SB431542 |
3 days + 25 days | D14: 16% PECAM1+ | N/A | [29] |
F | rEC (early vs. late) | Human dermal fibroblast | ETV2 | VEGFA and VPA | Lentiviral infection |
Early rEC: EGM-2 and VEGFA Late rEC: EGM-2, temporal treatment of VPA, and VEGFA |
7 days for early rEC and 3 months for late rEC | D7: 50% CDH5+, 39% KDR+ D93: 60% CDH5+, 83% PECAM1 + | HLI model LDPI (4 weeks) IHC: Early rEC (4 weeks) and Late rEC (3 months) | [30] |
G | EiEC | Human adipose- derived stem cell and human umbilical mesenchymal stem cells | ETV2 | SB431542, CHIR99021, VEGF, bFGF, EGF, and BMP4 | Lentiviral infection |
EIM: Insulin, ascorbic acid, Heparin, VEGF, bFGF, EGF, SB431542, CHIR99021 and BMP4 EMM: SB431542, VEGF, bFGF, and EGF |
10 days(up to 2 months) |
D10: 47.2% KDR+ D30: 73.8% PECAM1+, 60.6% KDR+, 88.1% CDH5+ | HLI Model (2 weeks) Injected with 30% Matrigel | [35] |
H | iEC | Human embryonic lung fibroblast | DKK3 | VEGF | Adenoviral infection | EGM-2 and VEGF | 10 days or more | N/A | Matrigel plug (1 week) | [31] |
I | Fsk-iEC | Human fibroblast and UCB-MSC | ETV2 | forskolin | Lentiviral or Retroviral infection | EGM-2 and Forskolin | 14 days | D14: 55.9% CDH5+, 43.9% PECAM1+ | Matrigel plug (1 week) HLI model (2 weeks) | [33] |
J | iVEC | Human dermal fibroblast | ETV2 | Retroviral infection | EGM-2 MV | N/A | N/A | N/A | [32] | |
K | rCVT | Mouse tail-tip fibroblast | miR-208b- 3p | ascorbic acid and BMP4 | Lipid-based transfection | DMEM/F-12, 10% FBS, ascorbic acid, and BMP4 | 10 days | D10: 28% PECAM1+ | MI model Masson’s trichrome staining and Echocardiography (12 weeks) IHC (16 weeks) | [46] |
L | iEC | Human adult dermal fibroblast | ETV2, KLF2 and T AL1 with siTWIST 1 | Rosiglitazone | Lentiviral infection |
1st stage: EGM-2 MV and rosiglitazone 2nd stage: EGM-2 MV |
4 weeks + 2 weeks | D28: 63% CDH5+, 9.5% PECAM1 + D42: 19.6% PECAM1+ and CDH5+ | N/A | [34] |
M | SCAP-EC* | Human MSC from apical papilla | VPA, CHIR99021, Repsox, Forskolin, Y- 27632 | Chemical stimulation |
1 st Induction Medium: EGM-2, VEGF and BMP4 2nd Induction Medium: EGM-2, VEGF, and 8- Br-3,5-cAMP |
4 days + 4 days | D8: 42.88% KDR+, 39.20% TEK+, 14.10% PECAM1 + | Matrigel plug (2 weeks) | [37] | |
N | ETV2 overexpressing DPSC | Dental pulp stem cells | ETV2 | Lentiviral infection | Differentiation media: EGM | 14 days | D7: 69.9% CDH5 + | Matrigel plug (1 week) | [36] |
DPSC = dental pulp stem cell; EBM-2 = Endothelial Cell Growth Basal Medium-2; EC = endothelial cell; EGM-2 MV = Microvascular Endothelial Cell Growth Medium-2; EiEC = ETV2-induced endothelial cell; EIM = endothelial induction medium; EM = endothelial growth media; EMM = endothelial maintenance medium; Fsk = forskolin; HLI = hindlimb ischemia model; iEC = induced endothelial cell; iEnd cell = induced endothelial cell; IHC=immunohistochemistry; iVEC = induced vascular endothelial cell; LDPI = laser doppler perfusion imaging; N/A = not applicable; rAC-VEC = reprogrammed amniotic fluid-derived cell-vascular endothelial cell; rCVT = reprogrammed cardiovascular tissue; rEC = reprogrammed endothelial cell; SCAP-EC = stem cell from apical papilia-derived chemical-induced endothelial cells; siTWIST = small interfering RNA of TWIST1; UCB-MSC = umbilical cord blood-derived mesenchymal stem cell; VEGF = vascular endothelial growth factor; VPA = valproic acid.
Chemically driven direct reprogramming towards ECs.
2.2.1. Source cells for endothelial reprogramming.
Therapeutic application of direct reprogramming starts with the selection of appropriate source cells. An adequate number of source cells should be acquired in non-invasive and cost-effective manners. In addition, the success of direct reprogramming greatly depends on the source cells. It is important to distinguish the reprogrammability of each source as reprogramming efficiency may differ for many aspects such as species, cell types, and tissue of origin. For example, Han et al. successfully reprogrammed ECs from murine fibroblasts using Foxo1, Etv2, Klf2, Tal1, and Lmo2, but they failed to generate ECs from human bone marrow mononuclear cells with the five factors [25]. Ginsberg et al. were the first to demonstrate direct EC reprogramming from human cells [26]. They suggested human ACs as an ideal source amenable for reprogramming. The cells are highly proliferative, can be human leukocyte antigens (HLA)-typed, and are cryopreservable for clinical use. However, human ACs are not suitable for autologous cell therapy, and their reprogramming methods were not applicable for human adult somatic cells. They acknowledged that ACs express ETV2 cofactors FOXC2, which drives endothelial gene expression with FOX:ETS elements. Though they showed ACs did not express pluripotency markers like OCT4, SOX2, and NANOG, midgestation ACs possibly have more plastic chromatin state permissible for reprogramming. To date, the majority of studies gave attention to fibroblasts as the possible source cells applicable for autologous cell therapy [27–34]. Cheng et al. suggested hADSC, an easily accessible cell type with multipotentiality, as a candidate somatic cell source for autologous cell therapy [35]. They successfully generated ECs from hADSCs via ETV2 overexpression. Interestingly, Li et al. and Yi et al. used a unique type of stem or progenitor cells derived from dental tissue in their reprogramming studies [36, 37]. Li et al. overexpressed ETV2 to generate ECs from dental pulp stem cells (DPSCs) [36]. Yi et al. used chemicals and small molecules to convert mesenchymal stem cells from apical papilla (SCAPs) into ECs [37].
2.2.2. Reprogramming strategy using transcription factors.
In early 2010, direct conversion of somatic cells to other cell types without activating pluripotency was demonstrated in generating neuronal cells from mouse fibroblasts [38]. Since then, numerous studies attempted to directly reprogram various cell types with unique sets of TFs and culture conditions [39–42]. In 2012, Ginsberg et al. pioneered a direct EC reprogramming strategy [26]. They reprogrammed endothelial cells from human amniotic fluid-derived cells (ACs) by overexpressing ETV2, FLI1, and ERG1. They suggested temporal regulation of ETV2 expression and TGFβ inhibition, increasing reprogramming efficiency and maturity of induced ECs. ETV2 was induced up to day 14 while FLI1 and ERG1 were continually over expressed. They treated SB431542, small molecule inhibitor for TGFβ, up to day 21. The reprogrammed cells, called rAC-VECs, displayed mature EC characteristics and formed tubular structures in vitro and in vivo Matrigel assays. Two years later, Han and colleagues used Foxo1, Etv2, Klf2, Tal1, and Lmo2 to directly reprogram mouse skin fibroblasts from Tie2-GFP reporter mice into induced ECs (iECs) [25]. Tie2-GFP positive iECs had similar in vivo characteristics as primary ECs isolated from the mouse lung. In another study, Wong and Cooke screened transcription factors and directly reprogrammed ECs from human dermal fibroblasts (HDFs) with ETV2, FLI1, GATA2, and KLF4 [29]. They noted ETV2 is the most essential factor for inducing PECAM1+ cell generation.
Human postnatal cell reprogramming toward ECs were first demonstrated by two studies (Lee et al. and Morita et al.), which showed that ETV2 alone is sufficient for reprogramming ECs from HDFs [27, 30]. Morita et al. demonstrated that ETV2 coordinates with endogenous FOXC2 to promote the FLI1 and ERG1 expression essential for downstream EC generation [27]. They noted that basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF) supplements improve reprogramming efficiency. Though they suggested that transient expression of ETV2 is sufficient to maintain PECAM1+ population, the reprogrammed cells retained high ETV2 expression even after 50 days in culture. This phenotype could be a transient or transitional cell because ETV2 is minimally expressed in postnatal ECs, and sustained expression of ETV2 during embryogenesis leads to vascular abnormalities. Extreme overexpression of ETV2 forced expression of CDH5 and PECAM1, which are direct targets of ETV2. Thus, the selected population with ectopic expression of EC markers were named as ETVECs, which were failed to show the maturation process of ECs. Lee et al., on the other hand, emphasized that temporal expression of ETV2 is not only necessary for directly reprogramming toward ECs but is sufficient for maturation of reprogrammed EC by applying appropriate culture strategies [30]. They presented the reprogrammed cells in two different stages. After initial activation of ETV2, KDR+ cells were sorted at day 7. These sorted cells, referred to as early reprogrammed ECs (rECs), showed EC phenotypes while lacking some mature characteristics. However, these early rECs lost their EC phenotypes 30 days after continued culture. At day 30, transient re-introduction of ETV2 with angiogenic culture conditions with or without valproic acid (VPA) for another 2 months generated more mature ECs and was called late rECs.
A pivotal role of ETV2 for human endothelial reprogramming was demonstrated by many follow-up studies. Cheng et al. reported direct conversion of ECs from human adipose-derived stem cells (hADSCs) by transiently expressing ETV2 and inhibiting TGFβ signaling pathway [35]. More recently, Kim et al. explored molecular mechanisms underlying ETV2-mediated endothelial transdifferentiation via RNA-seq and ChIP-seq analyses. They noted cyclic AMP (cAMP), exchange protein directly activated by cAMP (EPAC) and RAP1 signaling pathway played essential roles in EC reprogramming, and treating forskolin, a cAMP signaling activator, effectively increased reprogramming efficiency [33]. Bersini et al. converted human fibroblasts from donors of different age groups and patients with Hutchinson-Gilford Progeria Syndrome (HGPS) into induced smooth muscle cells (iSMC) and induced vascular ECs (iVECs) by over expressing MYOCD or ETV2, respectively [32]. Han et al. used three TFs (ETV2, KLF2, TAL1) to generate iECs from HDFs [34]. They noted CDH5 single positive cells lacked EC functionality and retained source cell signatures. On the other hand, CDH5 and PECAM1 double positive cells formed capillary tubes in Matrigel, secreted nitrogen oxide, and clustered closer to human umbilical vein EC (HUVEC) based on the sequencing analysis. From the RNA-seq analysis, they identified TWIST1, an epithelial-mesenchymal transition (EMT) activator as a factor blocking reprogramming. Supplementing Rosiglitazone, an MET inhibitor, and knocking out TWIST1 with siRNA system improved double positive cell generation. They also pointed out an additional two- week culture after CDH5 sorting helps increase double positive iEC generation. On the other hand, another study demonstrated endothelial reprogramming without ETV2. Chen et al. used DKK3, a secreted glycoprotein, a member of the dickkopf family of Wnt inhibitors, to convert human embryonic lung fibroblasts into functional ECs. DKK3 gene expression level picked up 2 days after infection and returned back to baseline after day 7. Such transient activation of DKK3 was sufficient to induce KDR gene expression. They also demonstrated that DKK3 contributes to the mesenchymal-to-epithelial transition (MET) and promotes KDR+ EC generation by modulating the miR-125a-5p/Stat3 axis pathway [31].
2.2.3. Delivery of reprogramming factors.
2.2.3.1. Viral delivery method.
For therapeutic translation of direct reprogramming, surveying effective and clinically compatible modes of gene delivery is required. To date, the majority of reprogramming studies employ lentiviral or retroviral vectors, which are integrated into the host genome and induce ectopic expression of the target gene expression [25–27, 29, 30, 32–35]. Ginsberg et al. and Lee et al. employed a doxycycline-inducible expression system to control ETV2 gene expression [26, 30]. After doxycycline (DOX) treatment, ETV2 gene and protein levels were elevated and induced downstream EC gene expression. After withdrawing DOX, directly reprogrammed ECs displayed phenotypes similar to mature fetal and adult ECs as they had low ETV2 levels while maintaining EC features. Even though the lentiviral or retroviral method is very efficient in activating target gene expression, insertional mutations induced by these viral methods is the major drawback for clinical application. Thus, non-integrating methods of gene delivery using DNA viruses such as adenovirus and adeno-associated virus (AAV) is more desirable. To date, one study reported the application of an adenoviral vector to overexpress a non-TF gene, DKK3, to reprogram human embryonic lung fibroblasts [31]. We have been using adenoviruses to transduce ETV2 to reprogram human adult cells into rECs (unpublished data). There is currently no study using AAV to directly induce EC reprogramming, but feasibility was demonstrated in in-vivo hepatic reprogramming study in which hepatic TFs were delivered via AAV6 vectors [43].
2.2.3.2. Non-viral delivery method.
Alternative to the viral vectors, non-viral delivery systems have been developed to avoid issues associated with cytotoxicity. Multiple studies demonstrated non-integrating episomal vectors to transfer genes and induced cellular reprogramming. In 2008, the Yamanaka group repeatedly transfected two genetically engineered plasmids and successfully generated human iPSCs [44]. Yu et al. also generated iPSCs with a single transfection of episomal vectors and confirmed complete removal of vector sequences from the reprogrammed cells [45]. Similarly, Kurien et al. episomally delivered OCT4, SOX2, KLF4, LMYC, LIN28, p53 short hairpin RNA, miR302, and miR367 to establish intermediate partial pluripotent state [23]. Most recently, Cho et al. simultaneously reprogrammed cardiomyocytes (rCMs), smooth muscle cells (rSMCs), and rECs from mouse fibroblasts by transfecting synthetic miRNA mimics, miR-208b-3p, with Lipofectamine [46]. Altogether, studies clearly demonstrate the feasibility of non-integrative delivery methods in inducing cellular reprograming.
2.2.4. Reprogramming mechanisms and small molecules for endothelial reprogramming
Most studies identified ETV2 as an essential TF for direct endothelial reprogramming. ETV2, a member of the ETS TF family, has an indispensable role in vascular development during embryogenesis [47]. Between embryonic days 7.0 and 9.0 in mice, hemato-endothelial progenitors in the mesoderm exhibit transient expression of ETV2 [48, 49]. ETV2-deficient mice embryos are unable to form vasculature and die at an early gestational age due to complete absence of hemato-endothelial lineage development [49, 50]. Etv2 has shown to activate key transcriptional regulators like Ets1 and Fli1 for endothelial development, and Tal1 and Gata1 for erythropoietic development [51–56]. Sinha et al. uncovered that different threshold Etv2 expression is required for initiating these different lineage development [57]. Etv2 functions in a feedforward manner, directly inducing intermediate transcription factors such as Ets1 and Fli1. Additionally, it binds to the promoter of EC related genes like Flk1, Cdh5, and Pecam1 and regulates their gene expressions as previously known [49, 50, 58]. The authors noted that Fli1 and Ets1 are not dose sensitive, and decrease in Etv2 expression dose does not disrupt vascular development. On the other hand, Tal1 is dose sensitive, and changes in Etv2 expression interrupts erythropoietic development. Furthermore, ETV2 collaborates with various co-factors, such as FOXC2, GATA2, and OVOL2 to specify endothelial lineage transitions [58–60]. Recently, Gong et al. addressed mechanisms of direct endothelial reprogramming by ETV2 induction employing scRNA-seq analysis [61]. Using mouse embryonic fibroblasts (MEFs), they elucidated that, at the initial step, ETV2 binds and recruits BRG1, an ATPase component of the SWI/SNF chromatin remodeling complex, onto closed chromatin domains to open chromatin of endothelial genes, as a pioneer factor. They also showed that BRG1 is required to maintain open states of chromatin by ETV2 binding. In another study, Kim et al. revealed that ETV2 targets RAP1 signaling. RAP1, a key regulator for integrin- and cadherin-based cell adhesion system, has an essential role in angiogenesis and vessel stabilization [62]. RAP1 is a downstream effector of cAMP/EPAC1 signaling, and supplementation of 8-Br-cAMP and forskolin, a cAMP analog and a cAMP inducer respectively, has shown to foster ETV2-mediated reprogramming with cAMP/EPAC/RAP1 signaling [33].
There are several studies, reporting the engagement of MET during the EC direct reprogramming process. Chen et al. reported that direct reprogramming of human embryonic lung fibroblasts into ECs by overexpression of DKK3 and found that a conversion process involves MET and miR-125a-5p/Stat3 signaling axis [31]. Han et al. showed inhibition of EMT with siTWIST and Rosiglitazone enhances the conversion efficiency [34]. Likewise, inhibition of TGFβ signaling cascade with SB431542 prevents EMT and helps maintain endothelial features [26].
Various combinations of growth factors, cytokines, and small molecules have been investigated to enhance reprogramming efficiency. Especially, EC growth factors like VEGF and bFGF have been commonly used. VEGF plays a vital role in EC differentiation by promoting the expression of EC-specific proteins [63–65]. bFGF signaling plays an important role in controlling endothelial homeostasis and maintaining EC fate [66, 67]. BMP4, a member of a TGFβ super family, was also used frequently to promote mesodermal lineage specification [66]. Lee et al. showed that VPA, a HDAC inhibitor, plays an additive role in enhancing EC reprogramming [30].
While many studies used TFs as primary reprogramming inducers, Sayed et al. used only small molecule compounds to convert HDFs into ECs without overexpressing TFs [28]. They first activated innate immunity with Poly I:C, a toll like receptor 3 (TLR3) agonist, for 7 days. They cultured the cells in a transdifferentiation media containing VEGF, bFGF, and BMP4 for another week. Then, they added 8-Br-cAMP in the media and cultured for 2 weeks. Lastly, they sorted the PECAM1+ cells and maintained the cells in the EGM-2 media with SB431542. The study suggests innate immunity is crucial for epigenetic plasticity and provides an environment suitable for EC transdifferentiation. Recently, Yi et al. used VPA, CHIR99021, an inhibitor of GSK3 resulting in activation of the WNT pathway, Repsox, a TGFβ signaling inhibitor suppressing EMT, Forskolin, an activator of cAMP pathway, and Y-27632 to convert SCAP into ECs [37]. Y-27632 is a Rho-associated protein kinase (ROCK) inhibitor shown to improve differentiation and expansion of ECs generated from embryonic-derived Flk1+ mesodermal precursor cells [68]. In addition to these 5 factors, they cultured the cells with VEGF, BMP4, and 8-Br-cAMP. The SCAP derived ECs had enriched endothelial gene expression and formed tubular structure both in vitro and in vivo Matrigel assays.
2.2.5. Role of reprogrammed endothelial cells in therapeutic neovascularization
2.2.5.1. Neovascularization capacity of reprogrammed ECs in non-disease model.
Clinical translation of directly reprogrammed ECs requires thorough investigation of their therapeutic potential and neovascularization capacity. A majority of studies used Matrigel to demonstrate functionality of their reprogrammed cells in vitro and in vivo [25–31, 33–37]. Distinctively, Chen et al. and Kim et al. assessed neovascularization capacity of directly reprogrammed ECs utilizing tissue engineered vascular grafts [31, 33]. Chen et al. seeded iECs in a decellularized mouse aortic graft with an ex vivo circulation bioreactor system [31]. The resultant iECs-reconstructed tissue graft displayed iECs lining the inner most layer surrounded by multiple SMC layers mimicking native vasculature. Kim et al. seeded KDR sorted iECs to an acellular rat liver scaffold and cultured the scaffold with a perfusion bioreactor system [33]. DOX and forskolin supplements in the perfusion medium promoted iECs to form confluent endothelium surrounding lumen of the scaffold as confirmed with immunostaining with Griffonia (Bandeiraea) Simplicifolia Lectin I (GSL I or BSL I), CDH5, and CLDN5. Ginsberg et al. demonstrated engraftment of rAV-VECs into the liver sinusoidal vessels of the NSG mice undergone 70% partial hepatectomy [26]. Three months after transplanting rAV-VECs into the intrasplenic route, they identified colocalization of their reprogrammed cells into the isolectin B4 perfused vessels. Though above studies demonstrated the potential of the reprogrammed ECs for various therapeutic applications, these models and studies did not directly address the therapeutic potential of directly reprogrammed ECs for ischemic cardiovascular diseases.
2.2.5.2. Contribution of reprogrammed ECs in therapeutic neovascularization.
Hindlimb ischemia (HLI) model has been used most commonly to investigate therapeutic effects of directly reprogrammed ECs. Han et al. intramuscularly injected iECs to the ischemic hindlimb and monitored blood flow recovery with laser Doppler perfusion imaging (LDPI) over 2 weeks [25]. iEC-injected group demonstrated blood flow recovery, vascular density, and tissue protective effects similar to the primary EC-injected group. The histological examination of the thigh muscle revealed that iECs were incorporated into the host vasculature labeled with BSL I. Sayed et al. injected their reprogrammed cells intramuscularly on the gastrocnemius muscle at two different time points [28]. First injection was done immediately after HLI induction. Second injection was done at 10 days after surgery when the signaling from bioluminescent labeled iECs became undetectable. They monitored blood flow recovery over 2 weeks with LDPI, and then they assessed degree of tissue damage and quantified blood capillary density on day 18 post-surgery. iEC injected group minimized tissue damage, toe necrosis at most, and improved blood flow in the ischemic hindlimbs similar to the human lung microvascular endothelial cells injected group. iEC-injected group had significantly higher capillary density compared to control group. Morita et al. demonstrated that ETVECs enhanced blood flow recovery and protected ischemic hindlimb from necrosis at day 14 after the cells were intramuscular injected into the adductor muscle immediately after HLI surgery [27]. Though, longer follow-ups for histological analysis were done only in Matrigel plug extracts. At day 28 after Matrigel plug implantation, they found colocalization of ETVECs, co-labeled by Ulexeuropaeus agglutinin I (UEA I) and CD34, with the CDH5+ host blood vessels. At day 42, ETVEC-constituting vessels were surrounded by ACTA2+ mural cells and expressed endothelial nitric oxide synthase (eNOS) indicating formation of mature and functional vessels. Similarly, Cheng et al. injected ETV2-induced endothelial-like cells (EiECs) suspended in 30% Matrigel intramuscularly on the adductor muscle immediately after HLI surgery [35]. EiECs injected group had improved blood perfusion and minimum tissue damage similar to the HUVEC injected group. In 14-day hindlimb tissue, EiECs constituting vessels were successfully stained with vascular markers like CDH5, PECAM1 and VWF, and had ACTA2+ mural cells surrounding them. Kim et al. collaterally injected forskolin-treated and KDR-sorted iECs (Fsk-iECs) from proximal to distal transected sites immediately after femoral artery ligation [33]. The Fsk-iECs treated group displayed blood flow recovery, tissue protective effect, and vessel density similar to HUVEC treated group. Lee et al. most clearly assessed therapeutic effects and neovascularization capacity of human directly reprogrammed EC [30]. They showed that both early and late rECs generated at two different stages of reprogramming have robust neovascularization potential in HLI tissue. Early rECs demonstrated better tissue protective effect, improved blood flow recovery, and higher capillary density compared to HUVECs over 4 weeks post-surgery. They were able to identify incorporation of early and late rEC into the host vasculature at 4 weeks and 3 months post-surgery.
While above studies presented promising therapeutic and neovascularization potential of direct reprogrammed ECs, long-term fate of the reprogrammed cells in vivo disease models are not well investigated. Han et al. showed that around 9% of the transplanted cells were engrafted in the ischemic tissue at 14 days after surgery even though the reprogrammed cells were generated from same murine origin [25]. Lee et al. reported around 7% of rECs were incorporated or anastomosed to the host vessels in the ischemic hindlimb tissue 28 days after surgery [30]. Recently, Cho et al. simultaneously generated cardiovascular tissue (rCVT) composed of rCMs, rSMCs and rECs with extracellular matrix via a direct reprogramming method [46]. They used miR-208b-3p, ascorbic acid, and BMP4 to directly convert mouse tail-tip fibroblasts into the tissue. Reprogrammed GFP labeled rCVT was transplanted onto the infarcted mouse heart. rCVT improved cardiac function, protected host myocardium from fibrosis, and reduced left ventricular wall thinning and compensatory enlargement of ventricles. They reported that rCVT-derived rECs migrated into the host infarcted heart and contributed to new vessel formation. Sixteen weeks after transplantation, 21% of vessels in the infarcted area were composed of or coaptated with GFP+ BSL I+ rECs, and GFP+ ACTA2+ reprogrammed SMCs ensheathed over the vessels composed of GFP− PECAM1+ host ECs and GFP+ PECAM1+ rECs. Even though the extent to which rECs alone contribute to therapeutical neovascularization on the infarcted heart still remain unknown, it is clear that administration of the vessel-forming cells embedded within extracellular matrix improved long-term cell retention and contribution to durable vessel formation.
3. Challenges for clinical application of reprogrammed ECs
3.1. Source cells and their origins
Cell source is also a critical aspect to consider when translating rECs for therapeutic purposes. The early studies of EC direct reprogramming used mouse cells and human fetal and neonatal foreskin fibroblasts demonstrating the concept and feasibility of direct reprogramming. Thereafter, fibroblasts collected from various tissues and subjects with different physiological conditions have been tested to systematically generate reprogrammed ECs. However, sourcing fibroblasts requires performing an invasive procedure and can potentially cause complications limiting multiple biopsies. When direct reprogramming aims for an autologous approach, the source cells will be collected from a patient. Collecting methods of source cells would better if they are less invasive and more reliable. For example, blood drawing for blood cell collection is less invasive compared to skin biopsy for skin fibroblasts collection. Even several groups studied cellular reprogramming of urine cells [69, 70] from collected urine, which is much easier and safer to collect compared to drawing blood and performing skin biopsies. The plasticity of cells is also an important element to consider. For EC direct reprogramming, mesodermal lineage cells such as smooth muscle cells and blood cells can be more easily reprogrammed. Not only lineage, but also the age of patients will affect the plasticity of source cells, which may eventually influence reprogramming efficiency. At present, the effect of the age of donor on reprogramming efficiency has been demonstrated in murine cells [71]. Meanwhile, studies on human cells have not consistently found a significant effect of aging on iPSC generation [72, 73]. However, Mertens et al. demonstrated that induced neurons generated from fibroblasts retained transcriptional aging signatures from aged donors [74]. Moreover, reprogramming efficiency has shown to be affected by cellular senescence [75, 76]. Since cellular senescence increases with donor age [77, 78], donor’s precondition may affect the reprogramming outcome. Thus, generally, reprogramming efficiency seems influenced by pre-existing signatures (or memories) of donor cells. In the case of direct EC reprogramming, although Bersini et al. did not find significant differences in reprogramming efficiency based on differential gene expression analysis, they reported heterogeneity in reprogrammed cell phenotypes caused by different ages and preconditions of patients [32]. iVECs from old donors had higher expression of GSTM1 and PALD1, which are associated with oxidative stress, inflammation, and vascular junction stability. In addition, BMPs overexpressed in iSMCs from HGPS were shown to be linked with endothelial barrier damage. This study indicates reprogrammed cells retain signature from original cell type and may affect their functionality.
3.2. Delivery of reprogramming factors
One of the main purpose of direct reprogramming is clinical application. As described in the earlier section, the previous studies demonstrated the potential of reprogrammed ECs as a source for cell therapy. However, when we considered the route of gene transduction or overexpression among previous studies, they are undesirable due to the integration of ectopic genes into host genomes by retroviral [23] or lentiviral [22, 24–27, 30, 79] vectors. These viral vectors are acceptable for biological studies or drug screenings but problematic for cell therapy. However, for clinical application, more favorable vectors for gene transduction should be developed for reprogrammed EC generation. For the choices of viral vectors, non-integrating properties should be considered first. The usage of non-integrating DNA viruses such as replication defective adenovirus or AAV have already been approved for gene therapy and adenovirus has been demonstrated for its feasibility for EC direct reprogramming by Chen et al. [31]. Sendai virus, a cytoplasmic RNA virus, has been shown to deliver transcription factors successfully for iPSC generation [80–82] and induction of cardiomyocytes [83] without the risk of genome integration. Owing to the non-integrating property, ectopic gene expression is not sustained with these viral vectors. Interestingly, the transient expression of transduced genes with these viral vectors is well suited for EC direct reprogramming using ETV2 because it is required in the early phase of reprogramming [26, 30].
Also, we can consider the direct delivery of genetic materials such as DNA in episomal vectors or modified mRNA. These methods are non-integrative and non-viral, and thus safe, clinically compatible, and able to keep host genome integrity. Gene expression through the introduction of DNA plasmids is a classical method of ectopic gene expression. Some early studies of iPSC generation [44, 45] and EC reprogramming via partial pluripotent state [23] used episomal vectors harboring pluripotency transcription factors suggesting that episomal vectors are capable of induce cellular reprogramming. However, when used for direct reprogramming, efficiency via this method is challenging. Modified mRNAs were used to replace viral vectors for iPSC generation a decade ago [84–86]. They were produced in in vitro transcription systems as possessing modified 5’-cap structure and nucleosides to evade the innate immunity and for better stability and translational efficiency. Later, direct reprogramming studies adopted modified mRNA to convert human fibroblasts into myoblast-like cells [87] and hepatocyte-like cells [88]. Although the feasibility of modified mRNA has been proved through these studies, mRNA methods have an advantage and a disadvantage over episomal vectors: its short half-life makes it free from vector clearance problem unlike episomal vectors, whereas mRNA needs to be transduced multiple rounds to maintain its expression at the desired level. Both mRNA and episomal vectors require aids of mechanical or chemical methods such as electroporation and transfection reagents for efficient delivery of genetic materials. Therefore, technical development for nucleic acid transfection must precede the clinical application to ensure cell viability and to maintain the expression of transcription factors, for example, synthetic self-replicative RNAs [89] and self-assembled mRNA nanoparticles [90].
3.3. Reprogramming efficiency
For clinical application, reprogramming efficiency is also important. Although iPSC and direct reprogramming strategies share similar concept of ectopically expressing specific transcription factors, iPSCs have the capacity of clonal expansion to produce an unlimited number of cells even from a single colony, whereas cells produced through direct reprogramming are somatic cells with limited capacity of proliferation. Therefore, the efficiency of direct reprogramming is much more critical. Aforementioned studies attempted to increase reprogramming efficiency employing additional reprogramming factors including novel transcription factors, signaling molecules to activate or inhibit specific signaling pathways, non-coding RNAs or epigenetic modulators. However, the most efficient method of reprogramming is unclear, due to different markers used to define reprogrammed cells and the varying time points at which the cells were collected. Han et al. was the first to make a direct comparison of the reprogramming efficiency between their method and methods from Ginsberg et al., Morita et al., Wong et al., and Lee et al. [34]. They derived rECs from human fibroblasts and noted that the proportion of CDH5/PECAM1 double-positive populations at D42 stayed below 15% regardless of which methods were used.
3.4. Subtypes of ECs
Furthermore, there should be an investigation of different phenotypes within reprogrammed cells to construct a better clinical model for subtype-specific cell therapy. Currently, there is not a clear understanding of how different endothelial subtypes can be generated from direct reprogramming strategies. During endothelial lineage development, arterial and venous endothelium are emerged from primitive vasculature. COUP-TFII has shown to play a critical role in venous development, and the knockdown of the TF resulted in inappropriate activation of arterial markers like Nrp1 and Notch1 in veins [91, 92]. You et al. suggested that COUP-TFII is a key factor for venous lineage specification, and the down regulation of Nrp1 expression inhibits Notch signaling and subsequent suppression of arterial genes [92]. Conversely, Hey1 and Hey2 are important modulators for Notch signaling, and knockdown of Hey genes suppressed expression of arterial markers while expression of venous markers increased [93, 94]. So far, studies used notch signaling inhibitor, such as γ-secretase inhibitor L-685, 458 (GSI), or agonist, such as resveratrol, to promote venous or arterial ECs differentiation from PSCs, respectively [95, 96]. Several studies also suggested that VEGF specify EC fate by binding at VEGFR2. High concentration of VEGF induced arterial-specification, and low concentration of VEGF induced venous-specification. Lymphatic ECs are another endothelial subtype that originates from venous endothelium. Prox1 has shown to be a master regulator for lymphatic development [97]. Prox1 knockdown caused failure in developing lymphatic vasculature [98], and overexpression of Prox1 increased expression of lymphatic genes like, Podoplanin and Vegfr3, while downregulating blood endothelial genes [99, 100]. Sox18 also has been considered as important factor that acts upstream of Prox1 [101]. Sox18 knockdown mice died with severe edema, while overexpression of Sox18 increased expression of lymphatic EC specific markers including Prox1. Lymphatic endothelial cells were successfully generated from iPSC with VEGF-C supplement [102]. Overall, an understanding of the transcription factors, growth factors, and signaling pathways involved in the specification of different endothelial subtypes can provide valuable insights for the development of direct reprogramming methods aimed at generating specific endothelial subtypes. Recent advance in sequencing technology for transcriptome, epigenome, proteome, and metabolome will provide clues to decipher the reprogramming mechanisms and to identify candidates for additional factors for reprogramming. Also, recent efforts to elucidate the heterogeneity of ECs with scRNA-seq analysis will be able to identify unique transcriptional programs specific to each subtype of ECs, which can be harnessed to enhance efficiency by further specification of EC subtypes during EC direct reprogramming [103, 104].
4. Conclusions
We have reviewed the studies of EC generation using direct reprogramming strategies. These studies not only demonstrated the feasibility of direct reprogramming as a method to produce functional ECs but also paved a new path to tackle cardiovascular diseases. These reprogrammed ECs exhibited phenotypic and functional EC characteristics in vitro and in vivo. Also, they displayed therapeutic potential in cardiovascular animal models. In addition, simpler procedures and shorter processing time compared to the generation of PSC derived cells as well as reasonable conversion efficiency with viral transduction make direct reprogramming a promising option to acquire autologous ECs for treating CVD via cell therapy.
As a reprogramming agent, not only new viral vectors such as Sendai viruses, adenoviruses, or AAV are clinically applicable but modified mRNA could be a great option. Various source cells need be explored to find more reliable and feasible options. Information obtained from the mechanism studies with high throughput sequencing and bioinformatics, especially single cell sequencing, will contribute to the identification of novel factors to improve direct reprogramming. In addition to generating reprogrammed cells in vitro, this direct reprogramming concept can be applied to in vivo cells and tissues. While such approach was demonstrated in mouse cardiomyocyte and neurons using lineage tracing, no studies clearly demonstrated such possibility for endothelial cells [105–109]. Direct cellular reprogramming also shed new light on generating a cardiovascular tissue in vitro which include cardiac and vascular cells using microRNAs and chemicals [46]. Despite such advance, there remains multiple questions to be answered for clinical application such as the long-term fate of reprogrammed cells and potential side effects. However, with rapid progress in this field, EC direct reprogramming strategy carries a great potential for treatment of cardiovascular diseases, disease modeling and drug discovery.
Highlights.
Cell therapy has been considered an attractive strategy to treat advanced ischemic cardiovascular diseases as it supplies cardiac and vascular components.
Direct reprogramming strategy, which aims to generate a target cell type using lineage-specific factors without undergoing pluripotency, received remarkable attentions for regenerative therapy.
Somatic cells from different origins have been successfully converted into functional endothelial cells with various direct reprogramming methods.
To enhance reprogramming efficiency, studies used combinations of various reprogramming factors such as transcription factors, angiogenic growth factors, and small molecular inhibitors or modulators for signaling pathways.
Acknowledgement
This work was supported by grants from NHLBI (R01HL150877, R01HL156008 and R01HL157242), AHA Career Development Award (19CDA34760061) and AHA Transformational Project Award (20TPA35490282), the Bio and Medical Technology Development Program of the National Research Foundation grant funded by the Korean government (MSIT) (2020M3A9I4038454, 2020R1A2C3003784) and the Faculty Research Assistance Program of Yonsei University College of Medicine for 2021 (6-2021-0178). Technical support for illustration was given by Hui Seon Cho.
Abbreviations
- AAV
adeno-associated viral
- AC
amniotic fluid-derived cell
- bFGF
basic fibroblast growth factor
- BMP4
bone morphogenetic protein 4
- BSL I
Griffonia (Bandeiraea) Simplicifolia Lectin I (GSL I)
- CM
cardiomyocyte
- CVD
cardiovascular disease
- DOX
doxycycline
- DPSCs
Dental pulp stem cells
- EC
endothelial cell
- EGM-2
Endothelial Cell Growth Media-2
- EiEC
ETV2-induced endothelial cell
- EMT
epithelial-to-mesenchymal transition
- eNOS
endothelial nitric oxide synthase
- ESC
embryonic stem cell
- Fsk
forskolin
- hADSCs
human adipose-derived stem cells
- HDF
human dermal fibroblasts
- HGPS
Hutchinson-Gilford Progeria Syndrome
- hiPSC
human induced pluripotent stem cell
- HLI
hindlimb ischemia model
- HUVEC
human umbilical vein EC
- iEC
induced endothelial cell
- iEnd
induced endothelial
- ILB4
isolectin B4
- iN
induced neuron
- iPSC
induced pluripotent stem cell
- iSMC
induced smooth muscle cell
- iVEC
induced vascular endothelial cell
- LDPI
laser Doppler perfusion imaging
- MET
mesenchymal-to-epithelial transition
- MI
myocardial infarction
- MIM
mesoderm induction medium
- miRNA
microRNA
- modRNA
modified mRNA
- OSKM
OCT4, SOX2, KLF4, c-MYC
- PiPSC
partial-induced pluripotent stem cell
- PSC
pluripotent stem cell
- rEC
reprogrammed endothelial cell
- rCVT
reprogrammed cardiovascular tissue
- SCAP
stem cells from apical papilla
- SCID
severe combined immunodeficiency
- shP53
p53 short hairpin RNA
- siRNA
small interfering RNA
- SMC
smooth muscle cell
- TGF
transforming growth factor
- TF
Transcription factor
- UBC-MSC
umbilical cord blood-derived mesenchymal stem cells
- UEA I
Ulexeuropaeus agglutinin
- VEGF
vascular endothelial growth factor
- VPA
valproic acid
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of Interest
Y-s Yoon is a co-founder of KarisBio Inc., but has no competing interests, as the work presented was performed independently. Other authors have no financial conflicts of interest.
References
- [1].Virani SS, Alonso A, Aparicio HJ, Benjamin EJ, Bittencourt MS, Callaway CW, Carson AP, Chamberlain AM, Cheng S, Delling FN, Elkind MSV, Evenson KR, Ferguson JF, Gupta DK, Khan SS, Kissela BM, Knutson KL, Lee CD, Lewis TT, Liu J, Loop MS, Lutsey PL, Ma J, Mackey J, Martin SS, Matchar DB, Mussolino ME, Navaneethan SD, Perak AM, Roth GA, Samad Z, Satou GM, Schroeder EB, Shah SH, Shay CM, Stokes A, VanWagner LB, Wang N-Y, Tsao CW, n. null, Heart Disease and Stroke Statistics—2021 Update, Circulation 143(8) (2021) e254–e743. [DOI] [PubMed] [Google Scholar]
- [2].Schillinger M, Sabeti S, Loewe C, Dick P, Amighi J, Mlekusch W, Schlager O, Cejna M, Lammer J, Minar E, Balloon Angioplasty versus Implantation of Nitinol Stents in the Superficial Femoral Artery, New England Journal of Medicine 354(18) (2006) 1879–1888. [DOI] [PubMed] [Google Scholar]
- [3].Serruys PW, Morice M-C, Kappetein AP, Colombo A, Holmes DR, Mack MJ, Ståhle E, Feldman TE, van den Brand M, Bass EJ, Van Dyck N, Leadley K, Dawkins KD, Mohr FW, Percutaneous Coronary Intervention versus Coronary-Artery Bypass Grafting for Severe Coronary Artery Disease, New England Journal of Medicine 360(10) (2009) 961–972. [DOI] [PubMed] [Google Scholar]
- [4].Weintraub WS, Grau-Sepulveda MV, Weiss JM, O’Brien SM, Peterson ED, Kolm P, Zhang Z, Klein LW, Shaw RE, McKay C, Ritzenthaler LL, Popma JJ, Messenger JC, Shahian DM, Grover FL, Mayer JE, Shewan CM, Garratt KN, Moussa ID, Dangas GD, Edwards FH, Comparative Effectiveness of Revascularization Strategies, New England Journal of Medicine 366(16) (2012) 1467–1476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Waldo SW, Secemsky EA, O’Brien C, Kennedy KF, Pomerantsev E, Sundt TM, McNulty EJ, Scirica BM, Yeh RW, Surgical Ineligibility and Mortality Among Patients With Unprotected Left Main or Multivessel Coronary Artery Disease Undergoing Percutaneous Coronary Intervention, Circulation 130(25) (2014) 2295–2301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [6].Farber A, Chronic Limb-Threatening Ischemia, New England Journal of Medicine 379(2) (2018) 171–180. [DOI] [PubMed] [Google Scholar]
- [7].Bhagra SK, Pettit S, Parameshwar J, Cardiac transplantation: indications, eligibility and current outcomes, Heart 105(3) (2019) 252. [DOI] [PubMed] [Google Scholar]
- [8].Warfarin I Antiplatelet Vascular Evaluation Trial, Anand S, Yusuf S, Xie C, Pogue J, Eikelboom J, Budaj A, Sussex B, Liu L, Guzman R, Cina C, Crowell R, Keltai M, Gosselin G, Oral anticoagulant and antiplatelet therapy and peripheral arterial disease, N Engl J Med 357(3) (2007) 217–27. [DOI] [PubMed] [Google Scholar]
- [9].Strauer BE, Brehm M, Zeus T, Köstering M, Hernandez A, Sorg RV, Kögler G, Wernet P, Repair of Infarcted Myocardium by Autologous Intracoronary Mononuclear Bone Marrow Cell Transplantation in Humans, Circulation 106(15) (2002) 1913–1918. [DOI] [PubMed] [Google Scholar]
- [10].Janssens S, Dubois C, Bogaert J, Theunissen K, Deroose C, Desmet W, Kalantzi M, Herbots L, Sinnaeve P, Dens J, Maertens J, Rademakers F, Dymarkowski S, Gheysens O, Van Cleemput J, Bormans G, Nuyts J, Belmans A, Mortelmans L, Boogaerts M, Van de Werf F, Autologous bone marrow-derived stem-cell transfer in patients with ST-segment elevation myocardial infarction: double-blind, randomised controlled trial, The Lancet 367(9505) (2006) 113–121. [DOI] [PubMed] [Google Scholar]
- [11].Lunde K, Solheim S, Aakhus S, Arnesen H, Abdelnoor M, Egeland T, Endresen K, Ilebekk A, Mangschau A, Fjeld JG, Smith HJ, Taraldsrud E, Grøgaard HK, Bjørnerheim R, Brekke M, Müller C, Hopp E, Ragnarsson A, Brinchmann JE, Forfang K, Intracoronary Injection of Mononuclear Bone Marrow Cells in Acute Myocardial Infarction, New England Journal of Medicine 355(12) (2006) 1199–1209. [DOI] [PubMed] [Google Scholar]
- [12].Meyer GP, Wollert KC, Lotz J, Pirr J, Rager U, Lippolt P, Hahn A, Fichtner S, Schaefer A, Arseniev L, Ganser A, Drexler H, Intracoronary bone marrow cell transfer after myocardial infarction: 5-year follow-up from the randomized-controlled BOOST trial, Eur Heart J 30(24) (2009) 2978–84. [DOI] [PubMed] [Google Scholar]
- [13].Takahashi K, Yamanaka S, Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors, Cell 126(4) (2006) 663–676. [DOI] [PubMed] [Google Scholar]
- [14].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 131(5) (2007) 861–872. [DOI] [PubMed] [Google Scholar]
- [15].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, Arteriosclerosis, Thrombosis, and Vascular Biology 29(7) (2009) 1100–1103. [DOI] [PubMed] [Google Scholar]
- [16].Oh JE, Jung C, Yoon Y.-s., Human Induced Pluripotent Stem Cell-Derived Vascular Cells: Recent Progress and Future Directions, Journal of Cardiovascular Development and Disease 8(11) (2021) 148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Rufaihah AJ, Huang NF, Jamé S, Lee JC, Nguyen HN, Byers B, De A, Okogbaa J, Rollins M, Reijo-Pera R, Gambhir SS, Cooke JP, Endothelial cells derived from human iPSCS increase capillary density and improve perfusion in a mouse model of peripheral arterial disease, Arterioscler Thromb Vasc Biol 31(11) (2011) e72–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Lee SJ, Sohn YD, Andukuri A, Kim S, Byun J, Han JW, Park IH, Jun HW, Yoon YS, Enhanced Therapeutic and Long-Term Dynamic Vascularization Effects of Human Pluripotent Stem Cell-Derived Endothelial Cells Encapsulated in a Nanomatrix Gel, Circulation 136(20) (2017) 1939–1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Cohen DE, Melton D, Turning straw into gold: directing cell fate for regenerative medicine, Nat Rev Genet 12(4) (2011) 243–52. [DOI] [PubMed] [Google Scholar]
- [20].Yamanaka S, A Fresh Look at iPS Cells, Cell 137(1) (2009) 13–17. [DOI] [PubMed] [Google Scholar]
- [21].Zhou Q, Brown J, Kanarek A, Rajagopal J, Melton DA, In vivo reprogramming of adult pancreatic exocrine cells to beta-cells, Nature 455(7213) (2008) 627–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Margariti A, Winkler B, Karamariti E, Zampetaki A, Tsai TN, Baban D, Ragoussis J, Huang Y, Han JD, Zeng L, Hu Y, Xu Q, Direct reprogramming of fibroblasts into endothelial cells capable of angiogenesis and reendothelialization in tissue-engineered vessels, Proc Natl Acad Sci U S A 109(34) (2012) 13793–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Kurian L, Sancho-Martinez I, Nivet E, Aguirre A, Moon K, Pendaries C, Volle-Challier C, Bono F, Herbert JM, Pulecio J, Xia Y, Li M, Montserrat N, Ruiz S, Dubova I, Rodriguez C, Denli AM, Boscolo FS, Thiagarajan RD, Gage FH, Loring JF, Laurent LC, Izpisua Belmonte JC, Conversion of human fibroblasts to angioblast-like progenitor cells, Nat Methods 10(1) (2013) 77–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Li J, Huang NF, Zou J, Laurent TJ, Lee JC, Okogbaa J, Cooke JP, Ding S, Conversion of human fibroblasts to functional endothelial cells by defined factors, Arterioscler Thromb Vasc Biol 33(6) (2013) 1366–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Han JK, Chang SH, Cho HJ, Choi SB, Ahn HS, Lee J, Jeong H, Youn SW, Lee HJ, Kwon YW, Cho HJ, Oh BH, Oettgen P, Park YB, Kim HS, Direct conversion of adult skin fibroblasts to endothelial cells by defined factors, Circulation 130(14) (2014) 1168–78. [DOI] [PubMed] [Google Scholar]
- [26].Ginsberg M, James D, Ding B-S, Nolan D, Geng F, Jason M Butler, Schachterle W, Venkat R. Pulijaal, Mathew S, Stephen T. Chasen, Xiang J, Rosenwaks Z, Shido K, Elemento O, Rabbany Sina Y., Rafii S, Efficient Direct Reprogramming of Mature Amniotic Cells into Endothelial Cells by ETS Factors and TGFβ Suppression, Cell 151(3) (2012) 559–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Morita R, Suzuki M, Kasahara H, Shimizu N, Shichita T, Sekiya T, Kimura A, K.-i. Sasaki, H. Yasukawa, A. Yoshimura, ETS transcription factor ETV2 directly converts human fibroblasts into functional endothelial cells, Proceedings of the National Academy of Sciences 112(1) (2015) 160–165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Sayed N, Wong WT, Ospino F, Meng S, Lee J, Jha A, Dexheimer P, Aronow BJ, Cooke JP, Transdifferentiation of Human Fibroblasts to Endothelial Cells, Circulation 131(3) (2015) 300–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Wong WT, Cooke JP, Therapeutic transdifferentiation of human fibroblasts into endothelial cells using forced expression of lineage-specific transcription factors, Journal of Tissue Engineering 7 (2016) 2041731416628329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Lee S, Park C, Han JW, Kim JY, Cho K, Kim EJ, Kim S, Lee S-J, Oh SY, Tanaka Y, Park I-H, An HJ, Shin CM, Sharma S, Y.-s. Yoon, Direct Reprogramming of Human Dermal Fibroblasts Into Endothelial Cells Using ER71/ETV2, Circulation Research 120(5) (2017) 848–861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Chen T, Karamariti E, Hong X, Deng J, Wu Y, Gu W, Simpson R, Wong MM, Yu B, Hu Y, Qu A, Xu Q, Zhang L, DKK3 (Dikkopf-3) Transdifferentiates Fibroblasts Into Functional Endothelial Cells—Brief Report, Arteriosclerosis, Thrombosis, and Vascular Biology 39(4) (2019) 765–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Bersini S, Schulte R, Huang L, Tsai H, Hetzer MW, Direct reprogramming of human smooth muscle and vascular endothelial cells reveals defects associated with aging and Hutchinson-Gilford progeria syndrome, eLife 9 (2020) e54383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Kim J-J, Kim D-H, Lee JY, Lee B-C, Kang I, Kook MG, Kong D, Choi SW, Woo H-M, Kim D-I, Kang K-S, cAMP/EPAC Signaling Enables ETV2 to Induce Endothelial Cells with High Angiogenesis Potential, Molecular Therapy 28(2) (2020) 466–478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Han J-K, Shin Y, Sohn M-H, Choi S-B, Shin D, You Y, Shin J-Y, Seo J-S, Kim H-S, Direct conversion of adult human fibroblasts into functional endothelial cells using defined factors, Biomaterials 272 (2021) 120781. [DOI] [PubMed] [Google Scholar]
- [35].Cheng F, Zhang Y, Wang Y, Jiang Q, Zhao C.j., Deng J, Chen X, Yao Y, Xia Z, Cheng L, Dai L, Shi G, Yang Y, Zhang S, Yu D, Wei Y, Deng H, Conversion of human adipose-derived stem cells into functional and expandable endothelial-like cells for cell-based therapies, Stem Cell Research & Therapy 9(1) (2018) 350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Li J, Zhu Y, Li N, Wu T, Zheng X, Heng B.c., Zou D, Xu J, Upregulation of ETV2 Expression Promotes Endothelial Differentiation of Human Dental Pulp Stem Cells, Cell Transplantation 30 (2021) 0963689720978739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Yi B, Ding T, Jiang S, Gong T, Chopra H, Sha O, Dissanayaka WL, Ge S, Zhang C, Conversion of stem cells from apical papilla into endothelial cells by small molecules and growth factors, Stem Cell Research & Therapy 12(1) (2021) 266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Vierbuchen T, Ostermeier A, Pang ZP, Kokubu Y, Südhof TC, Wernig M, Direct conversion of fibroblasts to functional neurons by defined factors, Nature 463(7284) (2010) 1035–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Ieda M, Fu JD, Delgado-Olguin P, Vedantham V, Hayashi Y, Bruneau BG, Srivastava D, Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors, Cell 142(3) (2010) 375–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Szabo E, Rampalli S, Risueño RM, Schnerch A, Mitchell R, Fiebig-Comyn A, Levadoux-Martin M, Bhatia M, Direct conversion of human fibroblasts to multilineage blood progenitors, Nature 468(7323) (2010) 521–6. [DOI] [PubMed] [Google Scholar]
- [41].Pang ZP, Yang N, Vierbuchen T, Ostermeier A, Fuentes DR, Yang TQ, Citri A, Sebastiano V, Marro S, Südhof TC, Wernig M, Induction of human neuronal cells by defined transcription factors, Nature 476(7359) (2011) 220–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [42].Sekiya S, Suzuki A, Direct conversion of mouse fibroblasts to hepatocyte-like cells by defined factors, Nature 475(7356) (2011) 390–3. [DOI] [PubMed] [Google Scholar]
- [43].Rezvani M, Español-Suñer R, Malato Y, Dumont L, Grimm AA, Kienle E, Bindman JG, Wiedtke E, Hsu BY, Naqvi SJ, Schwabe RF, Corvera CU, Grimm D, Willenbring H, In Vivo Hepatic Reprogramming of Myofibroblasts with AAV Vectors as a Therapeutic Strategy for Liver Fibrosis, Cell Stem Cell 18(6) (2016) 809–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Okita K, Nakagawa M, Hyenjong H, Ichisaka T, Yamanaka S, Generation of mouse induced pluripotent stem cells without viral vectors, Science 322(5903) (2008) 949–53. [DOI] [PubMed] [Google Scholar]
- [45].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 324(5928) (2009) 797–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Cho J, Kim S, Lee H, Rah W, Cho HC, Kim NK, Bae S, Shin DH, Lee MG, Park IH, Tanaka Y, Shin E, Yi H, Han JW, Hwang PTJ, Jun HW, Park HJ, Cho K, Lee SW, Jung JK, Levit RD, Sussman MA, Harvey RP, Yoon YS, Regeneration of infarcted mouse hearts by cardiovascular tissue formed via the direct reprogramming of mouse fibroblasts, Nat Biomed Eng 5(8) (2021) 880–896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Sumanas S, Lin S, Ets1-related protein is a key regulator of vasculogenesis in zebrafish, PLoS Biol 4(1) (2006) e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].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, Proceedings of the National Academy of Sciences 106(3) (2009) 814–819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Lee D, Park C, Lee H, Lugus JJ, Kim SH, Arentson E, Chung YS, Gomez G, Kyba M, Lin S, Janknecht R, Lim D-S, Choi K, ER71 Acts Downstream of BMP, Notch, and Wnt Signaling in Blood and Vessel Progenitor Specification, Cell Stem Cell 2(5) (2008) 497–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Liu F, Kang I, Park C, Chang L-W, Wang W, Lee D, Lim D-S, Vittet D, Nerbonne JM, Choi K, ER71 specifies Flk-1+ hemangiogenic mesoderm by inhibiting cardiac mesoderm and Wnt signaling, Blood 119(14) (2012) 3295–3305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Fujiwara Y, Browne CP, Cunniff K, Goff SC, Orkin SH, Arrested development of embryonic red cell precursors in mouse embryos lacking transcription factor GATA-1, Proceedings of the National Academy of Sciences 93(22) (1996) 12355–12358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Pevny L, Simon MC, Robertson E, Klein WH, Tsai S-F, D’Agati V, Orkin SH, Costantini F, Erythroid differentiation in chimaeric mice blocked by a targeted mutation in the gene for transcription factor GATA-1, Nature 349(6306) (1991) 257–260. [DOI] [PubMed] [Google Scholar]
- [53].Robb L, Lyons I, Li R, Hartley L, Köntgen F, Harvey RP, Metcalf D, Begley CG, Absence of yolk sac hematopoiesis from mice with a targeted disruption of the scl gene, Proceedings of the National Academy of Sciences 92(15) (1995) 7075–7079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Bloor AJC, Sánchez M-J, Green AR, Göttgens B, The Role of the Stem Cell Leukemia (SCL) Gene in Hematopoietic and Endothelial Lineage Specification, Journal of Hematotherapy & Stem Cell Research 11(2) (2002) 195–206. [DOI] [PubMed] [Google Scholar]
- [55].De Val S, Black BL, Transcriptional Control of Endothelial Cell Development, Developmental Cell 16(2) (2009) 180–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].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 111(9) (2008) 4500–4510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [57].Sinha T, Lammerts van Bueren K, Dickel DE, Zlatanova I, Thomas R, Lizama CO, Xu SM, Zovein AC, Ikegami K, Moskowitz IP, Pollard KS, Pennacchio LA, Black BL, Differential Etv2 threshold requirement for endothelial and erythropoietic development, Cell Reports 39(9) (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].De Val S, Chi NC, Meadows SM, Minovitsky S, Anderson JP, Harris IS, Ehlers ML, Agarwal P, Visel A, Xu S-M, Pennacchio LA, Dubchak I, Krieg PA, Stainier DYR, Black BL, Combinatorial Regulation of Endothelial Gene Expression by Ets and Forkhead Transcription Factors, Cell 135(6) (2008) 1053–1064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Shi X, Richard J, Zirbes KM, Gong W, Lin G, Kyba M, Thomson JA, Koyano-Nakagawa N, Garry DJ, Cooperative interaction of Etv2 and Gata2 regulates the development of endothelial and hematopoietic lineages, Developmental Biology 389(2) (2014) 208–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Kim JY, Lee RH, Kim TM, Kim D-W, Jeon Y-J, Huh S-H, Oh S-Y, Kyba M, Kataoka H, Choi K, Ornitz DM, Chae J-I, Park C, OVOL2 is a critical regulator of ER71/ETV2 in generating FLK1+, hematopoietic, and endothelial cells from embryonic stem cells, Blood 124(19) (2014) 2948–2952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Gong W, Das S, Sierra-Pagan JE, Skie E, Dsouza N, Larson TA, Garry MG, Luzete-Monteiro E, Zaret KS, Garry DJ, ETV2 functions as a pioneer factor to regulate and reprogram the endothelial lineage, Nat Cell Biol 24(5) (2022) 672–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Chrzanowska-Wodnicka M, Rap1 in endothelial biology, Curr Opin Hematol 24(3) (2017) 248–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Simons M, Eichmann A, Molecular controls of arterial morphogenesis, Circ Res 116(10) (2015) 1712–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Dejana E, Hirschi KK, Simons M, The molecular basis of endothelial cell plasticity, Nature Communications 8(1) (2017) 14361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Roca C, Adams RH, Regulation of vascular morphogenesis by Notch signaling, Genes Dev 21(20) (2007) 2511–24. [DOI] [PubMed] [Google Scholar]
- [66].Marcelo KL, Goldie LC, Hirschi KK, Regulation of Endothelial Cell Differentiation and Specification, Circulation Research 112(9) (2013) 1272–1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Chen P-Y, Qin L, Barnes C, Charisse K, Yi T, Zhang X, Ali R, Medina Pedro P., Yu J, Slack Frank J., Anderson Daniel G., Kotelianski V, Wang F, Tellides G, Simons M, FGF Regulates TGF-β Signaling and Endothelial-to-Mesenchymal Transition via Control of let-7 miRNA Expression, Cell Reports 2(6) (2012) 1684–1696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Joo HJ, Choi D-K, Lim JS, Park J-S, Lee S-H, Song S, Shin JH, Lim D-S, Kim I, Hwang K-C, Koh GY, ROCK suppression promotes differentiation and expansion of endothelial cells from embryonic stem cell–derived Flk1+ mesodermal precursor cells, Blood 120(13) (2012) 2733–2744. [DOI] [PubMed] [Google Scholar]
- [69].Wang L, Wang L, Huang W, Su H, Xue Y, Su Z, Liao B, Wang H, Bao X, Qin D, He J, Wu W, So KF, Pan G, Pei D, Generation of integration-free neural progenitor cells from cells in human urine, Nat Methods 10(1) (2013) 84–9. [DOI] [PubMed] [Google Scholar]
- [70].Zhou T, Benda C, Duzinger S, Huang Y, Li X, Li Y, Guo X, Cao G, Chen S, Hao L, Chan Y-C, Ng K-M, Cy Ho J, Wieser M, Wu J, Redl H, Tse H-F, Grillari J, Grillari-Voglauer R, Pei D, Esteban MA, Generation of Induced Pluripotent Stem Cells from Urine, Journal of the American Society of Nephrology 22(7) (2011) 1221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [71].Wang B, Miyagoe-Suzuki Y, Yada E, Ito N, Nishiyama T, Nakamura M, Ono Y, Motohashi N, Segawa M, Masuda S, Takeda S, Reprogramming efficiency and quality of induced Pluripotent Stem Cells (iPSCs) generated from muscle-derived fibroblasts of mdx mice at different ages, PLoS Curr 3 (2011) Rrn1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [72].Trokovic R, Weltner J, Noisa P, Raivio T, Otonkoski T, Combined negative effect of donor age and time in culture on the reprogramming efficiency into induced pluripotent stem cells, Stem Cell Research 15(1) (2015) 254–262. [DOI] [PubMed] [Google Scholar]
- [73].Lapasset L, Milhavet O, Prieur A, Besnard E, Babled A, Aït-Hamou N, Leschik J, Pellestor F, Ramirez JM, De Vos J, Lehmann S, Lemaitre JM, Rejuvenating senescent and centenarian human cells by reprogramming through the pluripotent state, Genes Dev 25(21) (2011) 2248–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [74].Mertens J, Paquola ACM, Ku M, Hatch E, Böhnke L, Ladjevardi S, McGrath S, Campbell B, Lee H, Herdy JR, Gonçalves JT, Toda T, Kim Y, Winkler J, Yao J, Hetzer MW, Gage FH, Directly Reprogrammed Human Neurons Retain Aging-Associated Transcriptomic Signatures and Reveal Age-Related Nucleocytoplasmic Defects, Cell Stem Cell 17(6) (2015) 705–718. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Li H, Collado M, Villasante A, Strati K, Ortega S, Cañamero M, Blasco MA, Serrano M, The Ink4/Arf locus is a barrier for iPS cell reprogramming, Nature 460(7259) (2009) 1136–1139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Banito A, Rashid ST, Acosta JC, Li S, Pereira CF, Geti I, Pinho S, Silva JC, Azuara V, Walsh M, Vallier L, Gil J, Senescence impairs successful reprogramming to pluripotent stem cells, Genes Dev 23(18) (2009) 2134–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].Campisi J, d’Adda di Fagagna F, Cellular senescence: when bad things happen to good cells, Nature Reviews Molecular Cell Biology 8(9) (2007) 729–740. [DOI] [PubMed] [Google Scholar]
- [78].Narita M, Nuñez S, Heard E, Narita M, Lin AW, Hearn SA, Spector DL, Hannon GJ, Lowe SW, Rb-Mediated Heterochromatin Formation and Silencing of E2F Target Genes during Cellular Senescence, Cell 113(6) (2003) 703–716. [DOI] [PubMed] [Google Scholar]
- [79].Hong X, Margariti A, Le Bras A, Jacquet L, Kong W, Hu Y, Xu Q, Transdifferentiated Human Vascular Smooth Muscle Cells are a New Potential Cell Source for Endothelial Regeneration, Sci Rep 7(1) (2017) 5590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Fusaki N, Ban H, Nishiyama A, Saeki K, Hasegawa M, Efficient induction of transgene-free human pluripotent stem cells using a vector based on Sendai virus, an RNA virus that does not integrate into the host genome, Proc Jpn Acad Ser B Phys Biol Sci 85(8) (2009) 34862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Seki T, Yuasa S, Oda M, Egashira T, Yae K, Kusumoto D, Nakata H, Tohyama S, Hashimoto H, Kodaira M, Okada Y, Seimiya H, Fusaki N, Hasegawa M, Fukuda K, Generation of induced pluripotent stem cells from human terminally differentiated circulating T cells, Cell Stem Cell 7(1) (2010) 11–4. [DOI] [PubMed] [Google Scholar]
- [82].Ban H, Nishishita N, Fusaki N, Tabata T, Saeki K, Shikamura M, Takada N, Inoue M, Hasegawa M, Kawamata S, Nishikawa S, Efficient generation of transgene-free human induced pluripotent stem cells (iPSCs) by temperature-sensitive Sendai virus vectors, Proc Natl Acad Sci U S A 108(34) (2011) 14234–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Miyamoto K, Akiyama M, Tamura F, Isomi M, Yamakawa H, Sadahiro T, Muraoka N, Kojima H, Haginiwa S, Kurotsu S, Tani H, Wang L, Qian L, Inoue M, Ide Y, Kurokawa J, Yamamoto T, Seki T, Aeba R, Yamagishi H, Fukuda K, Ieda M, Direct In Vivo Reprogramming with Sendai Virus Vectors Improves Cardiac Function after Myocardial Infarction, Cell Stem Cell 22(1) (2018) 91–103 e5. [DOI] [PubMed] [Google Scholar]
- [84].Plews JR, Li J, Jones M, Moore HD, Mason C, Andrews PW, Na J, Activation of pluripotency genes in human fibroblast cells by a novel mRNA based approach, PLoS One 5(12) (2010) e14397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Warren L, Manos PD, Ahfeldt T, Loh YH, Li H, Lau F, Ebina W, Mandal PK, Smith ZD, Meissner A, Daley GQ, Brack AS, Collins JJ, Cowan C, Schlaeger TM, Rossi DJ, Highly efficient reprogramming to pluripotency and directed differentiation of human cells with synthetic modified mRNA, Cell Stem Cell 7(5) (2010) 618–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Yakubov E, Rechavi G, Rozenblatt S, Givol D, Reprogramming of human fibroblasts to pluripotent stem cells using mRNA of four transcription factors, Biochem Biophys Res Commun 394(1) (2010) 189–93. [DOI] [PubMed] [Google Scholar]
- [87].Preskey D, Allison TF, Jones M, Mamchaoui K, Unger C, Synthetically modified mRNA for efficient and fast human iPS cell generation and direct transdifferentiation to myoblasts, Biochem Biophys Res Commun 473(3) (2016) 743–51. [DOI] [PubMed] [Google Scholar]
- [88].Simeonov KP, Uppal H, Direct reprogramming of human fibroblasts to hepatocyte-like cells by synthetic modified mRNAs, PLoS One 9(6) (2014) e100134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Yoshioka N, Gros E, Li HR, Kumar S, Deacon DC, Maron C, Muotri AR, Chi NC, Fu XD, Yu BD, Dowdy SF, Efficient generation of human iPSCs by a synthetic self-replicative RNA, Cell Stem Cell 13(2) (2013) 246–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Kim H, Park Y, Lee JB, Self-assembled Messenger RNA Nanoparticles (mRNA-NPs) for Efficient Gene Expression, Sci Rep 5 (2015) 12737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Pereira FA, Qiu Y, Zhou G, Tsai MJ, Tsai SY, The orphan nuclear receptor COUP-TFII is required for angiogenesis and heart development, Genes Dev 13(8) (1999) 1037–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].You L-R, Lin F-J, Lee CT, DeMayo FJ, Tsai M-J, Tsai SY, Suppression of Notch signalling by the COUP-TFII transcription factor regulates vein identity, Nature 435(7038) (2005) 98–104. [DOI] [PubMed] [Google Scholar]
- [93].Zhong TP, Childs S, Leu JP, Fishman MC, Gridlock signalling pathway fashions the first embryonic artery, Nature 414(6860) (2001) 216–220. [DOI] [PubMed] [Google Scholar]
- [94].Weinstein BM, Stemple DL, Driever W, Fishman MC, gridlock, a localized heritable vascular patterning defect in the zebrafish, Nature Medicine 1(11) (1995) 1143–1147. [DOI] [PubMed] [Google Scholar]
- [95].Ditadi A, Sturgeon CM, Tober J, Awong G, Kennedy M, Yzaguirre AD, Azzola L, Ng ES, Stanley EG, French DL, Cheng X, Gadue P, Speck NA, Elefanty AG, Keller G, Human definitive haemogenic endothelium and arterial vascular endothelium represent distinct lineages, Nature Cell Biology 17(5) (2015) 580–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Zhang J, Chu L-F, Hou Z, Schwartz MP, Hacker T, Vickerman V, Swanson S, Leng N, Nguyen BK, Elwell A, Bolin J, Brown ME, Stewart R, Burlingham WJ, Murphy WL, Thomson JA, Functional characterization of human pluripotent stem cell-derived arterial endothelial cells, Proceedings of the National Academy of Sciences 114(30) (2017) E6072–E6078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Adams RH, Alitalo K, Molecular regulation of angiogenesis and lymphangiogenesis, Nature Reviews Molecular Cell Biology 8(6) (2007) 464–478. [DOI] [PubMed] [Google Scholar]
- [98].Wigle JT, Oliver G, Prox1 Function Is Required for the Development of the Murine Lymphatic System, Cell 98(6) (1999) 769–778. [DOI] [PubMed] [Google Scholar]
- [99].Hong Y-K, Harvey N, Noh Y-H, Schacht V, Hirakawa S, Detmar M, Oliver G, Prox1 is a master control gene in the program specifying lymphatic endothelial cell fate, Developmental Dynamics 225(3) (2002) 351–357. [DOI] [PubMed] [Google Scholar]
- [100].Petrova TV, Mäkinen T, Mäkelä TP, Saarela J, Virtanen I, Ferrell RE, Finegold DN, Kerjaschki D, Ylä-Herttuala S, Alitalo K, Lymphatic endothelial reprogramming of vascular endothelial cells by the Prox-1 homeobox transcription factor, The EMBO Journal 21(17) (2002) 4593–4599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].François M, Caprini A, Hosking B, Orsenigo F, Wilhelm D, Browne C, Paavonen K, Karnezis T, Shayan R, Downes M, Davidson T, Tutt D, Cheah KSE, Stacker SA, Muscat GEO, Achen MG, Dejana E, Koopman P, Sox18 induces development of the lymphatic vasculature in mice, Nature 456(7222) (2008) 643–647. [DOI] [PubMed] [Google Scholar]
- [102].Lee S-J, Park C, Lee JY, Kim S, Kwon PJ, Kim W, Jeon YH, Lee E, Yoon Y.-s., Generation of pure lymphatic endothelial cells from human pluripotent stem cells and their therapeutic effects on wound repair, Scientific Reports 5(1) (2015) 11019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [103].Kalucka J, de Rooij LPMH, Goveia J, Rohlenova K, Dumas SJ, Meta E, Conchinha NV, Taverna F, Teuwen L-A, Veys K, García-Caballero M, Khan S, Geldhof V, Sokol L, Chen R, Treps L, Borri M, de Zeeuw P, Dubois C, Karakach TK, Falkenberg KD, Parys M, Yin X, Vinckier S, Du Y, Fenton RA, Schoonjans L, Dewerchin M, Eelen G, Thienpont B, Lin L, Bolund L, Li X, Luo Y, Carmeliet P, Single-Cell Transcriptome Atlas of Murine Endothelial Cells, Cell 180(4) (2020) 764–779.e20. [DOI] [PubMed] [Google Scholar]
- [104].Gong W, Das S, Sierra-Pagan JE, Skie E, Dsouza N, Larson TA, Garry MG, Luzete-Monteiro E, Zaret KS, Garry DJ, ETV2 functions as a pioneer factor to regulate and reprogram the endothelial lineage, Nature Cell Biology 24(5) (2022) 672–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Qian L, Huang Y, Spencer CI, Foley A, Vedantham V, Liu L, Conway SJ, Fu J.-d., Srivastava D, In vivo reprogramming of murine cardiac fibroblasts into induced cardiomyocytes, Nature 485(7400) (2012) 593–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Song K, Nam Y-J, Luo X, Qi X, Tan W, Huang GN, Acharya A, Smith CL, Tallquist MD, Neilson EG, Hill JA, Bassel-Duby R, Olson EN, Heart repair by reprogramming non-myocytes with cardiac transcription factors, Nature 485(7400) (2012) 599–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [107].Heinrich C, Bergami M, Gascón S, Lepier A, Viganò F, Dimou L, Sutor B, Berninger B, Götz M, Sox2-Mediated Conversion of NG2 Glia into Induced Neurons in the Injured Adult Cerebral Cortex, Stem Cell Reports 3(6) (2014) 1000–1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Niu W, Zang T, Smith Derek K., Vue Tou Y., Zou Y, Bachoo R, Johnson Jane E., Zhang C-L, SOX2 Reprograms Resident Astrocytes into Neural Progenitors in the Adult Brain, Stem Cell Reports 4(5) (2015) 780–794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Su Z, Niu W, Liu ML, Zou Y, Zhang CL, In vivo conversion of astrocytes to neurons in the injured adult spinal cord, Nat Commun 5 (2014) 3338. [DOI] [PMC free article] [PubMed] [Google Scholar]