Skip to main content

Sources and applications of endothelial seed cells: a review

Abstract

Endothelial cells (ECs) are widely used as donor cells in tissue engineering, organoid vascularization, and in vitro microvascular model development. ECs are invaluable tools for disease modeling and drug screening in fundamental research. When treating ischemic diseases, EC engraftment facilitates the restoration of damaged blood vessels, enhancing therapeutic outcomes. This article presents a comprehensive overview of the current sources of ECs, which encompass stem/progenitor cells, primary ECs, cell lineage conversion, and ECs derived from other cellular sources, provides insights into their characteristics, potential applications, discusses challenges, and explores strategies to mitigate these issues. The primary aim is to serve as a reference for selecting suitable EC sources for preclinical research and promote the translation of basic research into clinical applications.

Graphical abstract

Background

Endothelial cells (ECs) form a monolayer of flattened epithelial cells and are predominantly located on the inner luminal surface of blood vessels. ECs are widely distributed throughout the vascular system, which encompasses arteries, veins, and capillaries. ECs are pivotal as a hemostatic barrier, mediating blood–tissue interactions and securely maintaining vascular permeability and integrity through tight intercellular junctions. ECs actively secrete multifunctional bioactive molecules, including nitric oxide, angiotensin, prostaglandins, and thromboxanes, orchestrating processes such as vascular constriction, dilation, coagulation factor regulation, and immune response modulation. Furthermore, ECs regulate the metabolic homeostasis of the body’s three major nutrients—carbohydrates, proteins, and fats—thereby ensuring metabolic balance (Fig. 1).

Fig. 1
figure 1

Brief summary of EC functions. The functions include vascular wall formation, regulation of vascular permeability, inflammatory response mediation, thrombus formation inhibition, chemical signal secretion, regulation of vascular dilation and constriction. Figure created using BioRender.com

Researchers have extensively used the indispensable biological functions of ECs as seed cells for tissue-engineered vascular graft (TEVG) endothelialization and organoid model vascularization in preclinical investigations. Vascular transplantation is an effective approach for treating vascular diseases. Graft failure is primarily caused by inadequate endothelialization within the implanted scaffold, leading to thrombus formation and luminal obstruction. Consequently, deliberately integrating ECs significantly enhances vascular graft patency through augmented endothelialization [1]. Clinically relevant organoids undergo increased apoptosis as their volume increases primarily due to hypoxia and the accumulation of metabolic byproducts [2]. Introducing ECs to facilitate organoid vascularization improved blood perfusion and promoted more robust viability [2].

ECs are crucial in disease pathophysiology, including cardiovascular disorders, diabetes, chronic renal failure, and various tumors. These diseases can be influenced by specific pharmacological agents, such as antitumor agents, immunosuppressants, and growth factors, which modulate EC proliferation and migration. Consequently, they affect crucial processes such as angiogenesis and vascular repair. Harnessing the potential of ECs to establish in vitro disease models and drug screening platforms would enable the investigation of the underlying mechanisms of these diseases, conduct drug toxicity assessments, and foster the development of novel therapeutic agents in a controlled experimental environment. Several therapeutic approaches using endothelial progenitor cells (EPCs), particularly those enriched for the CD34 + and CD133 + phenotypes, have progressed to clinical trials [3]. These cell-based interventions have demonstrated promising effects in ameliorating angina symptoms, enhancing exercise tolerance, improving left ventricular function, and augmenting myocardial perfusion in patients with refractory angina, highlighting their potential in regenerative cardiovascular therapies.

Numerous EC sources are available for in vitro vascularization and laboratory research, and include stem cell-derived and primary cells. Nonetheless, a comprehensive summary and comparison of these cell sources is lacking. In this review, we attempt to present a comprehensive overview of the diverse sources of ECs, considering their applications, advantages, and disadvantages (Table 1). We aim to provide a reference to aid researchers in selecting the most appropriate EC sources for preclinical investigations, considering their specific research contexts.

Table 1 Brief summarization of different sources of ECs and their advantages, disadvantages and applications

Stem-cell derived ECs

Embryonic stem cells (ESCs)

A significant scientific milestone was achieved in 1998 when researchers successfully isolated ESCs from human blastocyst-stage embryos. This groundbreaking discovery unveiled the remarkable capacity of ESCs to differentiate into a wide array of cell types, including ECs [4]. Years of rigorous investigation established methodologies for ESC differentiation into ECs and are now widely used in scientific research.

The prominent ESC lines in use are the H1 and H9 lines. The induction strategies predominantly involve monolayer-directed differentiation into ECs and three-dimensional (3D) cultivation using embryoid bodies (EBs). Additionally, certain laboratories co-culture ESCs with stromal cells such as S17 mouse bone marrow (BM) cells, OP9 cells, and M2-10B4 cells to induce EC differentiation. Co-culture recreates a microenvironment that guides stem cell differentiation while avoiding the complexity of EBs. Nevertheless, co-culture may introduce xenogeneic elements, raising concerns about potential unknown biological contamination and immune rejection risk. Despite ongoing debates regarding the optimal method, each co-culture method successfully directed ESC differentiation into ECs with specific functional attributes. For example, human ESC-derived ECs (ESC-ECs) cultured through monolayer-directed differentiation produced nitric oxide, responded to chemical stimuli, and supported the development of new blood vessels, ultimately restarting blood flow in ischemic mouse limbs [5]. Shen et al. illustrated the potential of mouse ESC-ECs derived from EB formation for engineering blood vessels. That seminal study marked the initial evidence of ESCs as viable seed cells in tissue-engineered vascularization [6]. Furthermore, ESC-ECs represent a valuable cellular source for inducing vascular regeneration in ischemic myocardium tissue. When ESC-ECs cultured on fibronectin-coated dishes were introduced into the hearts of mice with ligated left anterior descending coronary arteries, microcapillaries and small veins were notably increased within the infarcted region, leading to improved functionality of the infarcted heart [7]. Moreover, ESC-ECs have been used under specific chemical conditions to construct versatile in vitro 3D vascular models [8] and were pivotal in establishing intricate vasculature-like networks in organoid systems [9,10,11]. These advancements have contributed significantly to the important preclinical research foundation required for drug screening, disease modeling, and related in vitro studies.

While ESC-ECs hold great promise for research, several issues should be considered: (1) the use and destruction of human embryos in ESC research result in religious and ethical concerns [12]. Recent Chinese policies have reinforced the alignment of ethical principles with social development, broadening the scope of patent protection for human ESC (hESC) research outcomes. This reinforcement has greatly encouraged technological advancements in this domain, fostering a conducive environment for innovation and discovery [13, 14]. (2) several readily available ESC lines are obtained through isolation and subsequent in vitro proliferation, often involving exposure to animal-derived components. This approach carries potential risks associated with toxic substances, microorganisms, and unforeseen biological contaminants, which may elicit immune responses. The strategies to address this issue include developing serum substitutes and culture medium free from harmful xenobiotics [15]. Additionally, implementing feeder-free culture systems is aimed at minimizing contamination from xenogeneic sources [16]; (3) transferring ESCs from embryos to culture dishes can lead to progressive adaptation to the new environmental conditions. This adaptation may result in alterations, including nuclear structure changes, DNA damage, double-strand DNA cleavage and fragmentation, loss of heterozygosity, increased cancer-related gene diversity [17], and the potential for mutations during extended cultivation periods [18]. Nevertheless, ESCs maintain a higher degree of genomic stability than somatic cells [19]. Genetic instability during ESC in vitro culture can be substantially reduced by optimizing cultivation conditions, following standardized protocols, minimizing passages, regular karyotyping, and vigilantly monitoring genomic stability. These strategies collectively aid the preservation of ESC integrity and reliability for research and potential clinical applications. (4) Somatic cell nuclear transfer (SCNT)-derived cells present a promising solution to immune rejection in transplantation, as they can be customized to match the patient’s genetic composition. Transferring the nucleus of a patient’s somatic cell into an enucleated oocyte creates ESCs (NT-ESCs) genetically identical to the patient. This method was successful in primates and is being adapted for use with human cells to produce stem cells compatible with the patient’s immune system for therapeutic purposes [20, 21].

Human induced pluripotent stem cells (hiPSCs)

The emergence of hiPSC technology has produced patient-specific hiPSCs free from ethical and immunological concerns [22]. hiPSC-derived ECs (hiPSC-ECs) that underwent CD144 antibody-conjugated magnetic bead sorting exhibited typical EC morphological and functional characteristics, including tubular structure formation, responsiveness to inflammatory signals, and nitric oxide production [23]. Notably, the hiPSC-ECs demonstrated an angiogenesis and endothelial differentiation capacity comparable to that of their ESC-derived counterparts. This capacity positions hiPSC-ECs as a valuable platform for disease modeling and drug screening [24].

The primary application of hiPSC-ECs is their direct use as a substitute for injured or dysfunctional ECs to facilitate impaired vascular system restoration. Transplanting hiPSC-ECs into an animal model of ischemia promoted the repair of injured tissues and enhanced tissue perfusion [25]. However, there is no definitive conclusion on the angiogenic capacity between hiPSC-ECs and human umbilical vein ECs (HUVECs). One study reported that hiPSC-ECs have a greater capacity to integrate into the developing zebrafish vascular system than HUVECs [26]. Conversely, other studies indicated that while hiPSC-ECs formed perfused vessels on the dorsal side of mice, they exhibited a reduced density and quantity of perfused vessels compared to HUVECs, resulting in diminished vessel maturation [27]. Nonetheless, these results collectively confirm the capacity of hiPSC-ECs to directly promote blood vessel formation in vivo and enhance blood perfusion for treating ischemic disorders.

hiPSC-ECs are pivotal in vascular engineering and organ transplantation. hiPSC-ECs are an abundant source of ECs and have wide-ranging applications in bioprinting [28], microfluidic devices [29], organ-on-chip technologies [30, 31], and organoid vascularization [32, 33]. Furthermore, hiPSC-ECs are a simplified yet potent platform for developing therapeutic interventions, enabling the accurate replication of human physiology that facilitates precise disease modeling and tissue regeneration.

The blood vessel organoids produced from hiPSC-ECs exhibited an impressive resemblance to the structure and function of native human blood vessels [34]. hiPSC-ECs implanted into decellularized vascular scaffolds demonstrated robust adhesion, stability, permeability, and sustained vascular patency [35]. Recent breakthroughs have emerged in utilizing hiPSC-ECs for constructing small-caliber TEVGs, where the implantation of these grafts in nude mice for 30 days revealed no noticeable thrombus formation, while non-endothelialized grafts exhibited significant thrombus formation [36]. These developments form a critical research foundation for developing innovative allogeneic vascular grafts for patients with vascular diseases.

Disease-specific hiPSC-ECs are a promising avenue for investigating differential gene expression. Using disease-specific hiPSCs avoids the constraints associated with gene editing and other requirements for disease modeling linked to ESCs. Disease-specific hiPSC-ECs have been successfully derived from patients with diabetes [37], peripheral artery disease [38], end-stage renal disease [39], type A hemophilia [40], and pulmonary arterial hypertension [41]. These disease-specific hiPSC-ECs facilitated the establishment of in vitro disease models with impaired endothelial function, providing crucial insights for developing therapeutic strategies based on autologous cells. Additionally, hiPSC-ECs from patients with genetic disorders and rare diseases exhibit disease-relevant phenotypic traits, providing vital preclinical data to clarify disease mechanisms and explore potential therapeutic approaches [42,43,44,45].

hiPSCs exhibit exceptional pluripotency compared to other cell types. hiPSCs exposed to specific tissues generated tissue-specific microvascular ECs (MVECs). Notably, hiPSC-derived brain MVECs could be integrated with organ-on-chip technology to create sophisticated neurovascular units [46]. This innovation enables the study of the blood–brain barrier response to relevant diseases and drug screening in the central nervous system [47, 48]. In a co-differentiation system involving hiPSC-ECs and hiPSC-derived cardiomyocytes, the hiPSC-ECs demonstrated cardiac marker expression similar to that observed in primary cardiac microvasculature [49]. This distinct characteristic confers hiPSC-ECs with the capacity to faithfully replicate the authentic biological state of ECs across multiple organs.

While reprogramming hiPSCs from patient or donor somatic cells is relatively straightforward, hiPSC-ECs have limited practical application in autologous cell therapy for elderly people or patients with genetic diseases. Gene editing technologies such as CRISPR can correct gene diseases in hiPSCs and efficiently model gene mutations [50,51,52]. Furthermore, hiPSCs carry the risk of tumorigenicity during differentiation. Accordingly, the evaluation of hiPSC-EC safety and stability can be facilitated by extending the culture duration to mimic senescence and by conducting in vivo tumorigenicity tests. Notably, even autologous hiPSCs exhibit a low immunogenicity profile [53]. Nevertheless, this concern can be addressed using human-derived components [54] or serum-free cultivation strategies [55] to avoid the use of xenogeneic materials in generating xeno-free (XF)-hiPSC-ECs.

Mesenchymal stem cells (MSCs)

MSCs represent a diverse subset of stromal stem cells with multipotent differentiation capacity that can be isolated from various human tissues, such as BM, adipose tissue (AT), umbilical cord (UC), UC blood, placenta, dental pulp (DP), and amniotic fluid [56]. MSCs from different anatomical sources exhibit a distinct potential for endothelial differentiation. For example, BM-MSCs isolated through density gradient centrifugation and stimulated with growth factors differentiated into ECs and formed capillary-like networks on matrix gels [57].

Laboratory research principally explores the therapeutic potential of MSC-ECs for ischemic diseases. For example, MSCs underwent differentiation into smooth muscle cells (SMCs) and ECs in a canine model of chronic ischemia, promoting angiogenesis and enhancing cardiac function [58]. Similarly, introducing MSCs into the endocardium in a porcine model of chronic myocardial infarction led to SMC and EC differentiation in the infarcted and border regions, facilitating the formation of blood vessels of varying sizes [59].

While MSCs are ubiquitously sourced, they exhibit heterogenous cellular morphology, surface marker expression, and differentiation potential, resulting in diverse biological characteristics across varying origins and batches. Furthermore, endothelial differentiation is typically subject to inefficiency and impurity. Specific growth factor cocktails, small molecules, or biomaterial scaffolds can augment differentiation efficacy and cell purity. Complementarily, advanced cell sorting techniques and functional assays ensure a homogenous and functional population of derived ECs.

Adult progenitor cell-derived ECs

Endothelial progenitor cells (EPCs)

In 1997, Asahara et al. [60] discovered circulating EPCs in peripheral blood, which were subsequently found to contribute significantly to vessel formation in murine and rabbit hindlimb ischemia models. These EPCs were characterized as a specific population of progenitor cells expressing markers such as CD133, CD34, and KDR. EPCs are EC precursor cells and are critical in angiogenesis and vasculogenesis [61]. EPCs are present in peripheral blood and have been identified in UC and BM. It was widely accepted that circulating EPCs originate from the BM. However, recent research using advanced techniques such as single-cell RNA sequencing, in vivo genetic tracing in murine models [62], and the analysis of ECs in the blood of allogeneic BM transplantation patients established that circulating EPCs do not originate from the BM [63]. Instead, they reside within the blood vessel walls.

TEVGs constructed using EPCs exhibit prolonged patency and demonstrate vascular contractile and relaxation characteristics similar to natural arteries [64]. Additionally, EPCs can swiftly coat implanted artificial vascular scaffolds, offering crucial support for in situ endothelialization induction [65]. Similar to MSCs, EPCs are also used in cell therapy for ischemic conditions. In vitro expanded EPCs enhanced cardiac function recovery, increased capillary density, reduced left ventricular scar formation, and facilitated vascular neogenesis in rat models of myocardial infarction [66]. Furthermore, the cells exhibited potential in ameliorating myocardial fibrosis, improving cardiac function in rat models with coronary microcirculatory disorders [67]. Combined transplantation is an alternative to the exclusive use of EPCs for therapeutic purposes. Co-transplanting EPCs and MSCs in laboratory settings, as opposed to EPCs alone, significantly enhanced organ function and fostered tissue regeneration and functional repair in ischemic diseases, albeit without significant differences in angiogenesis [68]. Therefore, the combined transplantation of EPCs and other stem cells holds potential advantages in tissue engineering. However, there is a pressing need for thorough foundational research to assess the feasibility of this approach before clinical trials can be initiated.

The isolation of EPCs from adult sources is hindered by their limited availability and challenges regarding isolation. Moreover, the discernible heterogeneity of EPCs, stemming from inconsistent culture conditions and a lack of standardization, has contributed to inconsistent experimental outcomes. These factors impose significant limitations on the widespread applicability of EPCs. Using more efficient and specific markers with advanced sorting technologies such as flow cytometry with multiple surface antigens (CD34 + , CD133 + , VEGFR2 +) enhances the purity and yield of the isolated EPCs. Establishing a unified, validated culture system and characterization criteria for EPCs fosters their self-renewal and preservation of characteristics, augmenting research outcome consistency and comparability.

Cardiac progenitor cells (CPCs)

In 2001, Orlic et al. successfully isolated c-Kit + cells from BM, which could differentiate into ECs and SMCs when exposed to cytokine mobilization. The CPCs subsequently migrated to the infarcted myocardium, contributing to myocardial cell regeneration [69]. Subsequent experiments validated the presence of CPCs in the heart. CPCs could differentiate into myocardial cells, SMCs, and ECs during cultivation. Additionally, CPCs were pivotal in promoting neovascularization within infarcted myocardium and facilitating cardiac function restoration. This groundbreaking discovery challenged the traditional notion that myocardial cells are terminally differentiated cells [70].

Various CPCs, including c-Kit + , Sca-1 + , Islet-1 + , cardiac side population, cardiospheres, and cardiosphere-derived cells, have been identified and extensively studied after isolation from the adult heart. In vivo, injected CPCs targeted the myocardial injury site and underwent in situ differentiation into cardiomyocytes, ECs, and SMCs [71, 72]. The intracoronary administration of c-Kit + CPCs in a rat model of acute myocardial infarction resulted in their division and differentiation into ECs and SMCs, effectively reducing the extent of myocardial infarction and enhancing cardiac function [73]. CPCs can be obtained from donor heart biopsies and cryopreserved cardiac tissues, and expanded and differentiated into ECs in vitro [74]. Nevertheless, CPC-ECs demonstrate limited utility compared to ECs derived from alternative sources in disease modeling and vascular engineering, suggesting a lack of superiority.

However, CPC acquisition is challenging due to their restricted availability. Additionally, cellular senescence and genetic mutations may arise during expansion and cultivation, affecting CPC quality and functionality. The complex task of characterizing and identifying CPCs is complicated by their phenotypic heterogeneity, which hinders the definition of specific markers for their isolation and purification. Hence, addressing these challenges and refining the procedures involved in CPC acquisition and cultivation are imperative to develop their therapeutic potential in regenerative medicine [75].

Primary ECs

HUVECs

In 1973, Jaffe et al. successfully isolated ECs from human umbilical veins and characterized them using morphological, immunohistochemical, and serological criteria [76]. Since then, HUVECs have been widely used by the global vascular biology community as they are easily obtained, their supply is abundant, and they are cost-effective. Importantly, the wealth of knowledge accumulated over the past five decades has firmly established HUVECs as the preferred source of ECs for scientific research. Additionally, HUVECs are the benchmark for developing innovative approaches in vascular biology and angiogenesis.

In 2004, Koike et al. [77] successfully co-cultured HUVECs with MSCs, constructing a vascular network in vitro. Upon transplantation into mice, this construct demonstrated a sustained presence for up to 1 year. Subsequently, as tissue engineering gained prominence, HUVECs have been extensively used as a source of ECs for tissue-engineered construct endothelialization [78, 79], vascular chip fabrication [80], bioink preparation [81], and various other applications. Data from 2013 to 2018 indicate that 59% of studies used HUVECs as the primary cell source for ECs [82]. Furthermore, HUVECs are used in organoid vascularization, aiding the development of functional blood vessel networks within these artificial organ-like structures. For example, Shi et al. co-cultured HUVECs with hESCs or hiPSCs, leading to their directed differentiation into vascularized brain organoids. HUVECs interconnected within the brain organoids formed a complex, permeable vascular system that persisted for > 200 days. Upon transplantation into animal models, HUVECs integrated with the host murine vascular ECs within the brain organoid, establishing a functional vascular network system characterized by blood flow. This integrated vascular system demonstrated the maturity and viability of the engrafted brain organoid [83]. Additionally, Takebe et al. co-cultured hiPSC-derived hepatic endoderm cells with HUVECs and BM-BMCs, inducing the formation of liver buds (hiPSC-LBs) comprising 3D spherical tissues. The hiPSC-LBs were transplanted into immunodeficient mice and generated intricate vascular networks within 48 h that integrated with the host vasculature. Real-time imaging confirmed the perfusion of host blood, validating the establishment of a functional human vascular network [84].

However, the application of HUVECs is subject to shortcomings. First, the inherent heterogeneity and tissue-specific nature of ECs in normal organs and tissues means that using HUVECs or other primary EC lines to generate vascular systems within organoids may not offer the same advantages as using PSC-derived ECs, which exhibit greater plasticity. Second, HUVECs have a limited lifespan, necessitating recurrent cell acquisition from fresh donors. Lastly, prolonged HUVEC culture may result in the loss of their native characteristics and functionality. Co-culturing HUVECs with ECs from other sources, such as specific adult or fetal tissues, and using 3D culture systems or bio-printing techniques to create microenvironments can better mimic in vivo conditions, enhancing the maintenance of HUVEC native morphology and function.

Other primary ECs

No discernable distinctions have been observed in cell proliferation, metabolic activity, membrane integrity, and vasoactive substance production between human umbilical artery-derived ECs (HUAECs) and HUVECs [85]. Consequently, some research laboratories use HUAECs as a source of ECs for in vitro investigations [86,87,88]. Primary ECs are also widely derived from various human tissues, extending beyond UC origins. MVECs isolated from the retina [89], AECs [90], coronary artery ECs [91], brain MVECs [92], and lung MVECs [93] have been used to promote vascularization in engineered tissues and organoids. Notably, several laboratories have devised methodologies for isolating MVECs from AT, which is abundant and accessible. However, differentiated MVECs constitute a heterogeneous amalgamation encompassing subpopulations with arterial, venous, and lymphatic lineages. Consequently, additional research is warranted to clarify the phenotypic heterogeneity of MVECs [94, 95]. These primary cells are a potential invaluable source for patient-specific cell therapies and in vitro disease modeling. However, their limited lifespan and relatively restricted adaptability limit their application. Currently, no compelling evidence supports their distinct advantage over HUVECs.

Lineage conversion ECs

Recent cellular reprogramming advancements have introduced a novel approach for converting one somatic cell type into another, bypassing the pluripotent state. This method involves inducing functional cells from a specific lineage through the exclusive use of lineage-restricted transcription factors, offering a promising means of generating functional ECs. Significantly, this strategy avoids the potential tumorigenic risks associated with PSC-derived ECs.

ETV2 is a pivotal factor in EC lineage regulation and reprogramming [96]. The transient expression of ETV2 enables the reprogramming of amniotic cells [97, 98] and human fibroblasts [99,100,101] into functional ECs without undergoing a pluripotent transition. These reprogrammed cells harbor the potential to establish mature and functional vascular systems in vivo, promoting blood flow restoration in ischemic limbs. Palikuqi et al. [102] “reset” mature human ECs into adaptive angiogenic cells through the transient expression of ETV2, which confers plasticity upon the ECs, allowing them to adapt to novel environments. Beyond ETV2, transducing Oct4, Klf4 [103] or Foxo1, Er71, Klf2, Tal1, and Lmo2 [104] into human fibroblasts induced their differentiation into functional ECs. Sayed et al. reported that innate immune signaling activation, achieved through the synergistic action of small molecules and Toll-like receptor 3 agonists combined with endothelial growth factors, successfully initiated human fibroblast transdifferentiation into ECs [105].

The donors’ age and health status can influence the state of reprogrammed ECs, encouraging efforts to leverage cell-regulating factors to enhance reprogramming.

Other cell-derived ECs

BM stromal cell subpopulations

BM-derived multipotent adult progenitor cells (MAPCs) can differentiate into ECs. MAPCs promoted angiogenesis through endothelial activation in a murine hindlimb ischemia model [106, 107]. While MAPCs and MSCs can be extracted from BM, multiple studies have conclusively demonstrated that MAPCs have significantly greater differentiation potential than MSCs [108, 109]. For example, MSC-like cells lacking Oct4 expression exhibit limited differentiation capabilities towards ECs and hepatocyte-like cells.

D’Ippolito et al. isolated a population of adult multipotent cells, termed marrow-isolated adult multilineage (MIAMI) cells, from the BM [110]. In vitro experiments established that the MIAMI cells could differentiate into CD31 + KDR + vWF + endothelial-like cells, which could form vascular network structures when cultured [111]. Furthermore, transplanting these cells into a murine hindlimb ischemia model facilitated the restoration of the ischemic region [112].

Kucia et al. [113] identified a population of very small embryonic-like stem cells (VSELs) in a subset of BM cells. Subsequently, at least 20 independent research groups confirmed the pluripotent differentiation potential of VSELs across germ layers. For example, transplanting human VSELs into an ischemic model demonstrated the emergence of CD31 + endothelial phenotypes [114]. Additionally, VSEL-like stem cells have been discovered in UC tissues [115]. However, no studies have demonstrated their capacity to regenerate 3D fully functional tissue structures or form teratomas in immunocompromised mice. Therefore, further experimental investigations are required to explore VSEL pluripotency [116].

In 2010, Kuroda et al. demonstrated that adult MSCs under in vitro stress conditions can undergo transformation, exhibiting pluripotent-like characteristics. They referred to these cells as Muse (multilineage differentiating stress-enduring) cells [117]. Following intravenous injection into a murine model of aortic aneurysm, Muse cells spontaneously differentiate into vascular SMCs and ECs, preserving the elastic fibers and attenuating aneurysm expansion [118]. Muse cells are believed to be abundantly distributed in the connective tissues of various human organs, demonstrating the capacity to differentiate into all three germ layers. Upon transplantation, Muse cells preferentially home in to injury sites and spontaneously differentiate into tissue-compatible cells. Due to their non-tumorigenic properties and exceptional homing capabilities, Muse cells are considered to have superior therapeutic potential compared to other types of stem cells [119, 120]. Despite their promising attributes, scientific understanding of Muse cells and their biological characteristics remains relatively limited, necessitating further exploration.

DP-derived ECs

Marchionni et al. successfully induced endothelial differentiation in DP stem cells (DPSCs) through exposure to VEGF. The induced DPSCs expressed Flt-1 and KDR and created tubular structures on a matrix [121]. Sakai et al. comprehensively investigated the differentiation potential of stem cells from human exfoliated deciduous teeth (SHED). Their work highlighted the successful differentiation of SHED into odontoblasts and ECs. Furthermore, SHED expressed key vascular markers through induction, including VEGFR2, CD31, and VE-cadherin, and effectively organized into functional capillary-like structures [122]. Nevertheless, the initial strategy for endothelial lineage differentiation exhibited remarkably low efficacy. To address this challenge, Gong et al. introduced an innovative approach that used the decellularized extracellular matrix from HUVECs. This bioactive substrate provided a favorable microenvironment for promoting SHED endothelial differentiation. Consequently, the mRNA expression levels of the endothelial-specific markers, CD31 and VEGFR-2, were significantly upregulated within the SHED population [123].

Overexpressing ETV2 in DPSCs promoted EC differentiation [124]. In the presence of small molecular compounds within the culture system, stem cells from apical papilla differentiated into EC-like cells while maintaining the tubular structure for a more extended period compared to the method proposed by Gong et al. [125]. However, the specific mechanisms underlying this phenomenon remain unexplored.

EC heterogeneity

Single-cell genomics and in vivo genetic labeling techniques have revealed the nuanced nature of ECs, which demonstrate organ specificity and heterogeneity during their developmental processes [126]. Different induction protocols or stages can yield diverse phenotypic traits within ECs. Consequently, each EC subgroup may be relevant in distinct research endeavors. For example, stem cells promoting angiogenesis, exemplified by stalk cells and tip cells, are ideal for exploring microvascularization in angiogenesis and tissue engineering. In contrast, phalanx ECs, characterized by weaker proliferative and migratory abilities, are more suitable for investigating arterial atherosclerosis and endothelialization in small-diameter vascular grafts [127]. Meticulous selection of the appropriate culture protocols and specific EC phenotypes for foundational research would enable the close replication of physiological or pathological conditions and yield reliable preclinical data.

Comparative characterization of ECs from various sources

Stem cell-derived ECs, notably iPSC-ECs and ESCs-ECs, exhibit robust proliferative capacity suitable for large-scale production and study, although iPSC-ECs demonstrate marginally reduced expression of key endothelial markers and shear-responsive genes compared to primary ECs [128]. iPSC-EC, ESCs-EC, and MSC proliferation, migration, and angiogenesis significantly outperform that of AECs [129, 130], yet the limited endothelial differentiation potential of MSCs renders them less ideal for vascular engineering. While iPSC-ECs form vessels with lower density and maturity than HUVECs in vasculogenesis [27, 131], they demonstrate excellent endothelial barrier function and responsiveness to shear stress [132].

Adult progenitor-derived ECs are hampered by restricted proliferation and differentiation abilities that may deteriorate with age [133]. Consequently, they are less effective in constructing vessels than HUVECs [134], primarily used in ischemic therapies in animal models.

Directly converting adult cells into ECs bypasses the pluripotent stage, simplifying the process and potentially offering better genetic stability with reduced mutation risks. However, this approach is subject to low conversion efficiency and functionality discrepancies with primary ECs, necessitating further conversion protocol optimization.

Conclusion and future perspectives

ECs are pivotal in the repair and regeneration of damaged tissues, demonstrating substantial potential in regenerative medicine. Investigating the origin of ECs is extremely important. This review examined the diverse origins of ECs used in preclinical research for clinical applications. HUVECs remain the predominant choice for in vitro EC research. However, the limited lifespan of HUVECs and primary ECs during cultivation and the potential loss of their phenotypic characteristics, suggest that more adaptable ECs from PSCs or progenitor cells may gradually supplant HUVECs. Alternatively, HUVECs could act exclusively as a reference standard for innovative vascular construction methods.

hESC-EC research has increased annually, and their application in basic research will become even more widespread following policy and regulation maturation. Contrastingly, hiPSCs present an unlimited cell source without harming human embryos while preserving the patient’s original disease-specific characteristics. Recent substantial progress has been made in using hiPSC-ECs for disease modeling and engineering tissue vascularization. Specifically, disease-specific iPSC-ECs can closely mimic vascular function and pathological features under disease conditions and will be predominantly used to provide innovative solutions to rare diseases or genetic vascular disorders, and used as drug screening platforms.

MSCs, EPCs, and CPCs exhibit immense potential in cell therapy for ischemic diseases. Their future applications will focus on providing effective support for recovery post-myocardial infarction, reducing the risk of heart failure.

Notably, attention to specific BM stromal cell subpopulations has increased. The capacity for differentiating into ECs and the broad clinical prospects thereof are currently in an intensive exploration phase. This field holds immense potential, heralding significant breakthroughs in regenerative medicine and disease therapeutics.

In summary, a comprehensive understanding of EC origins and characteristics is pivotal for the precise selection of regenerative medicine research models. This understanding would effectively facilitate the translation of research findings into clinical applications, expediting the development of personalized medicine and tissue repair technologies.

Abbreviations

3D:

Three-dimensional

AECs:

Aortic endothelial cells

AT:

Adipose tissue

CPCs:

Cardiac progenitor cells

BM:

Bone marrow

DP:

Dental pulp

DPSCs:

Dental pulp stem cells

EB:

Embryoid bodies

ECs:

Endothelial cells

EPCs:

Endothelial progenitor cells

ESCs:

Embryonic stem cells

ESC-ECs:

Embryonic stem cell-derived endothelial cells

hiPSCs:

Human induced pluripotent stem cells

hiPSC-ECs:

Human induced pluripotent stem cell-derived endothelial cells

hiPSC-LBs:

Human induced pluripotent stem cell-induced liver buds

HUAECs:

Human umbilical artery-derived endothelial cells

HUVECs:

Human umbilical vein endothelial cells

MAPCs:

Multipotent adult progenitor cells

MIAMI cells:

Marrow-isolated adult multilineage cells

MSCs:

Mesenchymal stem cells

Muse cells:

Multilineage differentiating stress-enduring cells

MVECs:

Microvascular endothelial cells

SHED:

Stem cells from human exfoliated deciduous teeth

SMCs:

Smooth muscle cells

TVEG:

Tissue engineered vascular graft

UC:

Umbilical cord

VSELs:

Very small embryonic-like stem cells

References

  1. Post A, Wang E, Cosgriff-Hernandez E. A review of integrin-mediated endothelial cell phenotype in the design of cardiovascular devices. Ann Biomed Eng. 2019;47(2):366–80.

    Article  PubMed  Google Scholar 

  2. Ryan AR, Cleaver O. Plumbing our organs: lessons from vascular development to instruct lab generated tissues. Curr Top Dev Biol. 2022;148:165–94.

    Article  CAS  PubMed  Google Scholar 

  3. Khan AR, Farid TA, Pathan A, Tripathi A, Ghafghazi S, Wysoczynski M, Bolli R. Impact of cell therapy on myocardial perfusion and cardiovascular outcomes in patients with angina refractory to medical therapy: a systematic review and meta-analysis. Circ Res. 2016;118(6):984–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Cyranoski D. How human embryonic stem cells sparked a revolution. Nature. 2018;555(7697):428–30.

    Article  CAS  PubMed  Google Scholar 

  5. Kane NM, Meloni M, Spencer HL, Craig MA, Strehl R, Milligan G, Houslay MD, Mountford JC, Emanueli C, Baker AH. Derivation of endothelial cells from human embryonic stem cells by directed differentiation: analysis of microRNA and angiogenesis in vitro and in vivo. Arterioscler Thromb Vasc Biol. 2010;30(7):1389–97.

    Article  CAS  PubMed  Google Scholar 

  6. Shen G, Tsung HC, Wu CF, Liu XY, Wang XY, Liu W, Cui L, Cao YL. Tissue engineering of blood vessels with endothelial cells differentiated from mouse embryonic stem cells. Cell Res. 2003;13(5):335–41.

    Article  PubMed  Google Scholar 

  7. Li Z, Wu JC, Sheikh AY, Kraft D, Cao F, Xie X, Patel M, Gambhir SS, Robbins RC, Cooke JP, et al. Differentiation, survival, and function of embryonic stem cell derived endothelial cells for ischemic heart disease. Circulation. 2007;116(11 Suppl):I46-54.

    PubMed  PubMed Central  Google Scholar 

  8. Sriram G, Handral HK, Gan SU, Islam I, Rufaihah AJ, Cao T. Fabrication of vascularized tissue constructs under chemically defined culture conditions. Biofabrication. 2020;12(4): 045015.

    Article  CAS  PubMed  Google Scholar 

  9. Cakir B, Xiang Y, Tanaka Y, Kural MH, Parent M, Kang YJ, Chapeton K, Patterson B, Yuan Y, He CS, et al. Engineering of human brain organoids with a functional vascular-like system. Nat Methods. 2019;16(11):1169–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Palakkan AA, Tarnick J, Waterfall M, Sallam M, Glykofrydis F, Elhendawi M, Davies JA. Production of kidney organoids arranged around single ureteric bud trees, and containing endogenous blood vessels, solely from embryonic stem cells. Sci Rep. 2022;12(1):12573.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Sun XY, Ju XC, Li Y, Zeng PM, Wu J, Zhou YY, Shen LB, Dong J, Chen YJ, Luo ZG. Generation of vascularized brain organoids to study neurovascular interactions. Elife. 2022; 11.

  12. Volarevic V, Markovic BS, Gazdic M, Volarevic A, Jovicic N, Arsenijevic N, Armstrong L, Djonov V, Lako M, Stojkovic M. Ethical and safety issues of stem cell-based therapy. Int J Med Sci. 2018;15(1):36–45.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Xie X, Chen J, Shu Z. From strict moral standards to ethical neutrality: a policy-guided shift in the patentability of human embryonic stem cells in China. Stem Cell Res Ther. 2020;11(1):499.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Peng YJ, Huang X, Zhou Q. Ethical and policy considerations for human embryo and stem cell research in China. Cell Stem Cell. 2020;27(4):511–4.

    Article  CAS  PubMed  Google Scholar 

  15. Stasi K, Goings D, Huang J, Herman L, Pinto F, Addis RC, Klein D, Massaro-Giordano G, Gearhart JD. Optimal isolation and xeno-free culture conditions for limbal stem cell function. Invest Ophthalmol Vis Sci. 2014;55(1):375–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Hannoun Z, Fletcher J, Greenhough S, Medine C, Samuel K, Sharma R, Pryde A, Black JR, Ross JA, Wilmut I, et al. The comparison between conditioned media and serum-free media in human embryonic stem cell culture and differentiation. Cell Reprogram. 2010;12(2):133–40.

    Article  CAS  PubMed  Google Scholar 

  17. Närvä E, Autio R, Rahkonen N, Kong L, Harrison N, Kitsberg D, Borghese L, Itskovitz-Eldor J, Rasool O, Dvorak P, et al. High-resolution DNA analysis of human embryonic stem cell lines reveals culture-induced copy number changes and loss of heterozygosity. Nat Biotechnol. 2010;28(4):371–7.

    Article  PubMed  Google Scholar 

  18. Lefort N, Feyeux M, Bas C, Féraud O, Bennaceur-Griscelli A, Tachdjian G, Peschanski M, Perrier AL. Human embryonic stem cells reveal recurrent genomic instability at 20q11.21. Nat Biotechnol. 2008; 26(12):1364–6.

  19. Hong Y, Stambrook PJ. Restoration of an absent G1 arrest and protection from apoptosis in embryonic stem cells after ionizing radiation. Proc Natl Acad Sci U S A. 2004;101(40):14443–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chung YG, Seay M, Elsworth JD, Redmond DE. Generation of pluripotent stem cells using somatic cell nuclear transfer and induced pluripotent somatic cells from African green monkeys. Stem Cells Dev. 2020;29(19):1294–307.

    Article  CAS  PubMed  Google Scholar 

  21. Yamada M, Johannesson B, Sagi I, Burnett LC, Kort DH, Prosser RW, Paull D, Nestor MW, Freeby M, Greenberg E, et al. Human oocytes reprogram adult somatic nuclei of a type 1 diabetic to diploid pluripotent stem cells. Nature. 2014;510(7506):533–6.

    Article  CAS  PubMed  Google Scholar 

  22. 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(5):861–72.

    Article  CAS  PubMed  Google Scholar 

  23. Paik DT, Tian L, Lee J, Sayed N, Mishra R, Wu JC. Large-scale single-cell RNA-Seq identifies heterogeneous populations of human primary and induced pluripotent stem cell-derived endothelial cells. Circul Res. 2018; 123.

  24. van Duinen V, Stam W, Mulder E, Famili F, Reijerkerk A, Vulto P, Hankemeier T, van Zonneveld AJ. Robust and scalable angiogenesis assay of perfused 3D human ipsc-derived endothelium for anti-angiogenic drug screening. Int J Mol Sci. 2020;21(13):1–9.

    Google Scholar 

  25. Shen WC, Chou YH, Huang HP, Sheen JF, Hung SC, Chen HF. Induced pluripotent stem cell-derived endothelial progenitor cells attenuate ischemic acute kidney injury and cardiac dysfunction. Stem Cell Res Ther. 2018;9(1):344.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Orlova VV, Drabsch Y, Freund C, Petrus-Reurer S, van den Hil FE, Muenthaisong S, Dijke PT, Mummery CL. Functionality of endothelial cells and pericytes from human pluripotent stem cells demonstrated in cultured vascular plexus and zebrafish xenografts. Arterioscler Thromb Vasc Biol. 2014;34(1):177–86.

    Article  CAS  PubMed  Google Scholar 

  27. Bezenah JR, Rioja AY, Juliar B, Friend N, Putnam AJ. Assessing the ability of human endothelial cells derived from induced-pluripotent stem cells to form functional microvasculature in vivo. Biotechnol Bioeng. 2019;116(2):415–26.

    Article  CAS  PubMed  Google Scholar 

  28. Dogan L, Scheuring R, Wagner N, Ueda Y, Schmidt S, Wörsdörfer P, Groll J, Ergün S. Human iPSC-derived mesodermal progenitor cells preserve their vasculogenesis potential after extrusion and form hierarchically organized blood vessels. Biofabrication. 2021; 13(4).

  29. Natividad-Diaz SL, Browne S, Jha AK, Ma Z, Hossainy S, Kurokawa YK, George SC, Healy KE. A combined hiPSC-derived endothelial cell and in vitro microfluidic platform for assessing biomaterial-based angiogenesis. Biomaterials. 2019;194:73–83.

    Article  CAS  PubMed  Google Scholar 

  30. Mandrycky CJ, Howard CC, Rayner SG, Shin YJ, Zheng Y. Organ-on-a-chip systems for vascular biology. J Mol Cell Cardiol. 2021;159:1–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Vila Cuenca M, Cochrane A, van den Hil FE, de Vries AAF, Lesnik Oberstein SAJ, Mummery CL, Orlova VV. Engineered 3D vessel-on-chip using hiPSC-derived endothelial- and vascular smooth muscle cells. Stem Cell Reports. 2021;16(9):2159–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Low JH, Li P, Chew EGY, Zhou B, Suzuki K, Zhang T, Lian MM, Liu M, Aizawa E, Rodriguez Esteban C, et al. Generation of human PSC-derived kidney organoids with patterned nephron segments and a De Novo vascular network. Cell Stem Cell. 2019;25(3):373-87.e9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Schumacher A, Roumans N, Rademakers T, Joris V, Eischen-Loges MJ, van Griensven M, LaPointe VLS. Enhanced microvasculature formation and patterning in iPSC-derived kidney organoids cultured in physiological hypoxia. Front Bioeng Biotechnol. 2022;10: 860138.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Wimmer RA, Leopoldi A, Aichinger M, Kerjaschki D, Penninger JM. Generation of blood vessel organoids from human pluripotent stem cells. Nat Protoc. 2019;14(11):3082–100.

    Article  CAS  PubMed  Google Scholar 

  35. Gara E, Zucchelli E, Jakus Z, Harding SE, Merkely B, Mathe D, Szabo G, Radovits T, Foldes G. Building functional vascular grafts from human pluripotent stem cells-derived endothelial cells. Cytotherapy. 2020;22(5):S10.

    Article  Google Scholar 

  36. Luo J, Qin L, Park J, Kural MH, Huang Y, Shi X, Riaz M, Wang J, Ellis MW, Anderson CW, et al. Readily available tissue-engineered vascular grafts derived from human induced pluripotent stem cells. Circ Res. 2022;130(6):925–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Jiang B, Wang X, Bolanos NR, Ameer G. Modeling diabetic endothelial dysfunction with patient-derived induced pluripotent stem cells (iPSCS). Circulation. 2018; 138.

  38. Boonkaew B, Suwanpitak S, Pattanapanyasat K, Sermsathanasawadi N, Wattanapanitch M. Efficient generation of endothelial cells from induced pluripotent stem cells derived from a patient with peripheral arterial disease. Cell Tissue Res. 2022;388(1):89–104.

    Article  CAS  PubMed  Google Scholar 

  39. Kim KW, Shin YJ, Kim B-M, Cui S, Ko EJ, Lim SW, Yang CW, Chung BH. Modeling of endothelial cell dysfunction using human induced pluripotent stem cells derived from patients with end-stage renal disease. Kidney Res Clin Pract. 2021;40(4):698–711.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Rose M, Gao K, Cortez-Toledo E, Agu E, Hyllen AA, Conroy K, Pan G, Nolta JA, Wang A, Zhou P. Endothelial cells derived from patients’ induced pluripotent stem cells for sustained factor VIII delivery and the treatment of hemophilia A. Stem Cells Transl Med. 2020;9(6):686–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Gu M, Donato M, Guo M, Wary N, Miao Y, Mao S, Saito T, Otsuki S, Wang L, Harper RL et al. iPSC-endothelial cell phenotypic drug screening and in silico analyses identify tyrphostin-AG1296 for pulmonary arterial hypertension. Sci Transl Med. 2021; 13(592).

  42. Sayed N, Liu C, Ameen M, Himmati F, Zhang JZ, Khanamiri S, Moonen JR, Wnorowski A, Cheng L, Rhee JW et al. Clinical trial in a dish using iPSCs shows lovastatin improves endothelial dysfunction and cellular cross-talk in LMNA cardiomyopathy. Sci Transl Med. 2020; 12(554).

  43. Ma B, Li T, Li W, Yang H, Zeng Q, Pan Z, Wang K, Chen Q, Xiong C, Zhou Z. Patient-specific and gene-corrected induced pluripotent stem cell-derived endothelial cells elucidate single-cell phenotype of pulmonary veno-occlusive disease. Stem Cell Rep. 2022.

  44. Zhou D, Tan Y, Liu X, Tang L, Wang H, Shen J, Wang W, Zhuang L, Tao J, Su J, et al. Patient-specific iPSC-derived endothelial cells reveal aberrant p38 MAPK signaling in atypical hemolytic uremic syndrome. Stem Cell Reports. 2021;16(9):2305–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Matrone G, Thandavarayan RA, Walther BK, Meng S, Mojiri A, Cooke JP. Dysfunction of iPSC-derived endothelial cells in human Hutchinson-Gilford progeria syndrome. Cell Cycle. 2019;18(19):2495–508.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Vatine GD, Barrile R, Workman MJ, Sances S, Barriga BK, Rahnama M, Barthakur S, Kasendra M, Lucchesi C, Kerns J, et al. Human iPSC-derived blood-brain barrier chips enable disease modeling and personalized medicine applications. Cell Stem Cell. 2019;24(6):995-1005.e6.

    Article  CAS  PubMed  Google Scholar 

  47. Piantino M, Louis F, Shigemoto-Mogami Y, Kitamura K, Sato K, Yamaguchi T, Kawabata K, Yamamoto S, Iwasaki S, Hirabayashi H, et al. Brain microvascular endothelial cells derived from human induced pluripotent stem cells as in vitro model for assessing blood-brain barrier transferrin receptor-mediated transcytosis. Mater Today Bio. 2022;14: 100232.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Yan L, Moriarty RA, Stroka KM. Recent progress and new challenges in modeling of human pluripotent stem cell-derived blood-brain barrier. Theranostics. 2021;11(20):10148–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Giacomelli E, Bellin M, Sala L, van Meer BJ, Tertoolen LG, Orlova VV, Mummery CL. Three-dimensional cardiac microtissues composed of cardiomyocytes and endothelial cells co-differentiated from human pluripotent stem cells. Development. 2017;144(6):1008–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Liu N, Olson EN. CRISPR modeling and correction of cardiovascular disease. Circ Res. 2022;130(12):1827–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. De Masi C, Spitalieri P, Murdocca M, Novelli G, Sangiuolo F. Application of CRISPR/Cas9 to human-induced pluripotent stem cells: from gene editing to drug discovery. Hum Genom. 2020;14(1):25.

    Article  Google Scholar 

  52. Flahou C, Morishima T, Takizawa H, Sugimoto N. Fit-For-all iPSC-derived cell therapies and their evaluation in humanized mice with NK cell immunity. Front Immunol. 2021;12: 662360.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Mehler VJ, Burns CJ, Stauss H, Francis RJ, Moore ML. Human iPSC-derived neural crest stem cells exhibit low immunogenicity. Mol Ther Methods Clin Dev. 2020;16:161–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Luo J, Shi X, Lin Y, Yuan Y, Kural MH, Wang J, Ellis MW, Anderson CW, Zhang SM, Riaz M, et al. Efficient differentiation of human induced pluripotent stem cells into endothelial cells under xenogeneic-free conditions for vascular tissue engineering. Acta Biomater. 2021;119:184–96.

    Article  CAS  PubMed  Google Scholar 

  55. Hamad S, Derichsweiler D, Gaspar JA, Brockmeier K, Hescheler J, Sachinidis A, Pfannkuche KP. High-efficient serum-free differentiation of endothelial cells from human iPS cells. Stem Cell Res Ther. 2022;13(1):251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Nombela-Arrieta C, Ritz J, Silberstein LE. The elusive nature and function of mesenchymal stem cells. Nat Rev Mol Cell Biol. 2011;12(2):126–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wang C, Li Y, Yang M, Zou Y, Liu H, Liang Z, Yin Y, Niu G, Yan Z, Zhang B. Efficient differentiation of bone marrow mesenchymal stem cells into endothelial cells in vitro. Eur J Vasc Endovasc Surg. 2018;55(2):257–65.

    Article  PubMed  Google Scholar 

  58. Silva GV, Litovsky S, Assad JA, Sousa AL, Martin BJ, Vela D, Coulter SC, Lin J, Ober J, Vaughn WK, et al. Mesenchymal stem cells differentiate into an endothelial phenotype, enhance vascular density, and improve heart function in a canine chronic ischemia model. Circulation. 2005;111(2):150–6.

    Article  CAS  PubMed  Google Scholar 

  59. Quevedo HC, Hatzistergos KE, Oskouei BN, Feigenbaum GS, Rodriguez JE, Valdes D, Pattany PM, Zambrano JP, Hu Q, McNiece I, et al. Allogeneic mesenchymal stem cells restore cardiac function in chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc Natl Acad Sci USA. 2009;106(33):14022–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, Witzenbichler B, Schatteman G, Isner JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275(5302):964–7.

    Article  CAS  PubMed  Google Scholar 

  61. Peichev M, Naiyer AJ, Pereira D, Zhu Z, Lane WJ, Williams M, Oz MC, Hicklin DJ, Witte L, Moore MA, et al. Expression of VEGFR-2 and AC133 by circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood. 2000;95(3):952–8.

    Article  CAS  PubMed  Google Scholar 

  62. Zhang Y, Babczyk P, Pansky A, Kassack MU, Tobiasch E. P2 Receptors Influence hMSCs Differentiation towards Endothelial Cell and Smooth Muscle Cell Lineages. Int J Mol Sci. 2020; 21(17).

  63. Aquino JB, Sierra R, Montaldo LA. Diverse cellular origins of adult blood vascular endothelial cells. Dev Biol. 2021;477:117–32.

    Article  CAS  PubMed  Google Scholar 

  64. Kaushal S, Amiel GE, Guleserian KJ, Shapira OM, Perry T, Sutherland FW, Rabkin E, Moran AM, Schoen FJ, Atala A, et al. Functional small-diameter neovessels created using endothelial progenitor cells expanded ex vivo. Nat Med. 2001;7(9):1035–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Rafii S, Oz MC, Seldomridge JA, Ferris B, Asch AS, Nachman RL, Shapiro F, Rose EA, Levin HR. Characterization of hematopoietic cells arising on the textured surface of left ventricular assist devices. Ann Thorac Surg. 1995;60(6):1627–32.

    Article  CAS  PubMed  Google Scholar 

  66. Shudo Y, Goldstone AB, Cohen JE, Patel JB, Hopkins MS, Steele AN, Edwards BB, Kawamura M, Miyagawa S, Sawa Y, et al. Layered smooth muscle cell-endothelial progenitor cell sheets derived from the bone marrow augment postinfarction ventricular function. J Thorac Cardiovasc Surg. 2017;154(3):955–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Xue Y, Zhou B, Wu J, Miao G, Li K, Li S, Zhou J, Geng Y, Zhang P. Transplantation of endothelial progenitor cells in the treatment of coronary artery microembolism in rats. Cell Transp. 2020;29:963689720912688.

    Article  Google Scholar 

  68. Sun K, Zhou Z, Ju X, Zhou Y, Lan J, Chen D, Chen H, Liu M, Pang L. Combined transplantation of mesenchymal stem cells and endothelial progenitor cells for tissue engineering: a systematic review and meta-analysis. Stem Cell Res Ther. 2016;7(1):151.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Orlic D, Kajstura J, Chimenti S, Jakoniuk I, Anderson SM, Li B, Pickel J, McKay R, Nadal-Ginard B, Bodine DM, et al. Bone marrow cells regenerate infarcted myocardium. Nature. 2001;410(6829):701–5.

    Article  CAS  PubMed  Google Scholar 

  70. Beltrami AP, Barlucchi L, Torella D, Baker M, Limana F, Chimenti S, Kasahara H, Rota M, Musso E, Urbanek K, et al. Adult cardiac stem cells are multipotent and support myocardial regeneration. Cell. 2003;114(6):763–76.

    Article  CAS  PubMed  Google Scholar 

  71. Oyama T, Nagai T, Wada H, Naito AT, Matsuura K, Iwanaga K, Takahashi T, Goto M, Mikami Y, Yasuda N, et al. Cardiac side population cells have a potential to migrate and differentiate into cardiomyocytes in vitro and in vivo. J Cell Biol. 2007;176(3):329–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Yoon J, Choi SC, Park CY, Shim WJ, Lim DS. Cardiac side population cells exhibit endothelial differentiation potential. Exp Mol Med. 2007;39(5):653–62.

    Article  CAS  PubMed  Google Scholar 

  73. Tillmanns J, Rota M, Hosoda T, Misao Y, Esposito G, Gonzalez A, Vitale S, Parolin C, Yasuzawa-Amano S, Muraski J, et al. Formation of large coronary arteries by cardiac progenitor cells. Proc Natl Acad Sci USA. 2008;105(5):1668–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Le TYL, Pickett HA, dos Remedios CG, Barbaro PM, Kizana E, Chong JJH. Platelet-derived growth factor receptor-alpha expressing cardiac progenitor cells can be derived from previously cryopreserved human heart samples. Stem Cells Develop. 2018;27(3):184–98.

    Article  CAS  Google Scholar 

  75. Monsanto MM, White KS, Kim T, Wang BJ, Fisher K, Ilves K, Khalafalla FG, Casillas A, Broughton K, Mohsin S, et al. Concurrent isolation of 3 distinct cardiac stem cell populations from a single human heart biopsy. Circ Res. 2017;121(2):113–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Jaffe EA, Nachman RL, Becker CG, Minick CR. Culture of human endothelial cells derived from umbilical veins. Identification by morphologic and immunologic criteria. J Clin Invest. 1973; 52(11):2745–56.

  77. Koike N, Fukumura D, Gralla O, Au P, Schechner JS, Jain RK. Tissue engineering: creation of long-lasting blood vessels. Nature. 2004;428(6979):138–9.

    Article  CAS  PubMed  Google Scholar 

  78. Weymann A, Schmack B, Okada T, Soós P, Istók R, Radovits T, Straub B, Barnucz E, Loganathan S, Pätzold I, et al. Reendothelialization of human heart valve neoscaffolds using umbilical cord-derived endothelial cells. Circ J. 2013;77(1):207–16.

    Article  PubMed  Google Scholar 

  79. Baptista PM, Siddiqui MM, Lozier G, Rodriguez SR, Atala A, Soker S. The use of whole organ decellularization for the generation of a vascularized liver organoid. Hepatology. 2011;53(2):604–17.

    Article  CAS  PubMed  Google Scholar 

  80. Zhang B, Montgomery M, Chamberlain MD, Ogawa S, Korolj A, Pahnke A, Wells LA, Massé S, Kim J, Reis L, et al. Biodegradable scaffold with built-in vasculature for organ-on-a-chip engineering and direct surgical anastomosis. Nat Mater. 2016;15(6):669–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Faramarzi N, Yazdi IK, Nabavinia M, Gemma A, Fanelli A, Caizzone A, Ptaszek LM, Sinha I, Khademhosseini A, Ruskin JN, et al. Patient-specific bioinks for 3D bioprinting of tissue engineering scaffolds. Adv Healthc Mater. 2018;7(11): e1701347.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Wang K, Lin RZ, Melero-Martin JM. Bioengineering human vascular networks: trends and directions in endothelial and perivascular cell sources. Cell Mol Life Sci. 2019;76(3):421–39.

    Article  CAS  PubMed  Google Scholar 

  83. Shi Y, Sun L, Wang M, Liu J, Zhong S, Li R, Li P, Guo L, Fang A, Chen R, et al. Vascularized human cortical organoids (vOrganoids) model cortical development in vivo. PLoS Biol. 2020;18(5): e3000705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Takebe T, Sekine K, Enomura M, Koike H, Kimura M, Ogaeri T, Zhang RR, Ueno Y, Zheng YW, Koike N, et al. Vascularized and functional human liver from an iPSC-derived organ bud transplant. Nature. 2013;499(7459):481–4.

    Article  CAS  PubMed  Google Scholar 

  85. Lau S, Gossen M, Lendlein A, Jung F. Venous and arterial endothelial cells from human umbilical cords: potential cell sources for cardiovascular research. Int J Mol Sci. 2021; 22(2).

  86. Catar R, Chen L, Zhao H, Wu D, Kamhieh-Milz J, Lücht C, Zickler D, Krug AW, Ziegler CG, Morawietz H et al. Native and oxidized low-density lipoproteins increase the expression of the LDL receptor and the LOX-1 receptor, respectively, in arterial endothelial cells. Cells. 2022; 11(2).

  87. Xue J, Fan J, Li Y, Wu W, Yan Q, Zheng Q. ABCG1 attenuates oxidative stress induced by H(2)O(2) through the inhibition of NADPH oxidase and the upregulation of Nrf2-mediated antioxidant defense in endothelial cells. Anal Cell Pathol. 2020;2020:2095645.

    Article  Google Scholar 

  88. Schreiber K, Sciascia S, Wehrmann F, Weiß C, Leipe J, Krämer BK, Stach K. The effect of hydroxychloroquine on platelet activation in model experiments. J Thromb Thrombol. 2021;52(2):674–9.

    Article  CAS  Google Scholar 

  89. Yoshida T, Komaki M, Hattori H, Negishi J, Kishida A, Morita I, Abe M. Therapeutic angiogenesis by implantation of a capillary structure constituted of human adipose tissue microvascular endothelial cells. Arterioscler Thromb Vasc Biol. 2010;30(7):1300–6.

    Article  CAS  PubMed  Google Scholar 

  90. Sasagawa T, Shimizu T, Yamato M, Okano T. Expression profiles of angiogenesis-related proteins in prevascular three-dimensional tissues using cell-sheet engineering. Biomaterials. 2014;35(1):206–13.

    Article  CAS  PubMed  Google Scholar 

  91. Bhattacharyya A, Lin S, Sandig M, Mequanint K. Regulation of vascular smooth muscle cell phenotype in three-dimensional coculture system by Jagged1-selective Notch3 signaling. Tissue Eng Part A. 2014;20(7–8):1175–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Thomsen MS, Humle N, Hede E, Moos T, Burkhart A, Thomsen LB. The blood-brain barrier studied in vitro across species. PLoS ONE. 2021;16(3): e0236770.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Tan Q, Choi KM, Sicard D, Tschumperlin DJ. Human airway organoid engineering as a step toward lung regeneration and disease modeling. Biomaterials. 2017;113:118–32.

    Article  CAS  PubMed  Google Scholar 

  94. Ryu YH, Moon SH, Kim KJ, Jun YJ, Oh DY, Kim SH, Rhie JW. A novel hypothesis and characterization to isolate microvascular endothelial cells simultaneously with adipose-derived stem cells from the human adipose-derived stromal vascular fraction. Tissue Eng Regen Med. 2021;18(3):429–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Antonyshyn JA, Mazzoli V, McFadden MJ, Gramolini AO, Hofer SOP, Simmons CA, Santerre JP. Mitigating the non-specific uptake of immunomagnetic microparticles enables the extraction of endothelium from human fat. Commun Biol. 2021;4(1):1205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. 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. 2022;24(5):672–84.

    Article  CAS  PubMed  Google Scholar 

  97. Ginsberg M, James D, Ding BS, Nolan D, Geng F, Butler JM, Schachterle W, Pulijaal VR, Mathew S, Chasen ST, et al. Efficient direct reprogramming of mature amniotic cells into endothelial cells by ETS factors and TGFβ suppression. Cell. 2012;151(3):559–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Ginsberg M, Schachterle W, Shido K, Rafii S. Direct conversion of human amniotic cells into endothelial cells without transitioning through a pluripotent state. Nat Protoc. 2015;10(12):1975–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Morita R, Suzuki M, Kasahara H, Shimizu N, Shichita T, Sekiya T, Kimura A, Sasaki K, Yasukawa H, Yoshimura A. ETS transcription factor ETV2 directly converts human fibroblasts into functional endothelial cells. Proc Natl Acad Sci U S A. 2015;112(1):160–5.

    Article  CAS  PubMed  Google Scholar 

  100. Lee S, Park C, Han JW, Kim JY, Cho K, Kim EJ, Kim S, Lee SJ, Oh SY, Tanaka Y, et al. Direct reprogramming of human dermal fibroblasts into endothelial cells using ER71/ETV2. Circ Res. 2017;120(5):848–61.

    Article  CAS  PubMed  Google Scholar 

  101. Park SY, Lee H, Kwon YW, Park MR, Kim JH, Kim JB. Etv2- and Fli1-induced vascular progenitor cells enhance functional recovery in ischemic vascular disease model-brief report. Arterioscler Thromb Vasc Biol. 2020;40(4):e105–13.

    Article  CAS  PubMed  Google Scholar 

  102. Palikuqi B, Nguyen DT, Li G, Schreiner R, Pellegata AF, Liu Y, Redmond D, Geng F, Lin Y, Gómez-Salinero JM, et al. Adaptable haemodynamic endothelial cells for organogenesis and tumorigenesis. Nature. 2020;585(7825):426–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. 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. 2013;33(6):1366–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Han JK, Chang SH, Cho HJ, Choi SB, Ahn HS, Lee J, Jeong H, Youn SW, Lee HJ, Kwon YW, et al. Direct conversion of adult skin fibroblasts to endothelial cells by defined factors. Circulation. 2014;130(14):1168–78.

    Article  CAS  PubMed  Google Scholar 

  105. 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: role of innate immunity. Circulation. 2015;131(3):300–9.

    Article  CAS  PubMed  Google Scholar 

  106. Ryu JC, Davidson BP, Xie A, Qi Y, Zha D, Belcik JT, Caplan ES, Woda JM, Hedrick CC, Hanna RN, et al. Molecular imaging of the paracrine proangiogenic effects of progenitor cell therapy in limb ischemia. Circulation. 2013;127(6):710–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Zhang J, Cui Y, Li X, Xiao Y, Liu L, Jia F, He J, Xie X, Parthasarathy S, Hao H, et al. 5F peptide promotes endothelial differentiation of bone marrow stem cells through activation of ERK1/2 signaling. Eur J Pharmacol. 2020;876: 173051.

    Article  CAS  PubMed  Google Scholar 

  108. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, Ortiz-Gonzalez XR, Reyes M, Lenvik T, Lund T, Blackstad M, et al. Pluripotency of mesenchymal stem cells derived from adult marrow. Nature. 2002;418(6893):41–9.

    Article  CAS  PubMed  Google Scholar 

  109. Ulloa-Montoya F, Kidder BL, Pauwelyn KA, Chase LG, Luttun A, Crabbe A, Geraerts M, Sharov AA, Piao Y, Ko MS, et al. Comparative transcriptome analysis of embryonic and adult stem cells with extended and limited differentiation capacity. Genome Biol. 2007;8(8):R163.

    Article  PubMed  PubMed Central  Google Scholar 

  110. D’Ippolito G, Diabira S, Howard GA, Menei P, Roos BA, Schiller PC. Marrow-isolated adult multilineage inducible (MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation potential. J Cell Sci. 2004;117(Pt 14):2971–81.

    Article  PubMed  Google Scholar 

  111. Rahnemai-Azar A, D’Ippolito G, Gomez LA, Reiner T, Vazquez-Padron RI, Perez-Stable C, Roos BA, Pham SM, Schiller PC. Human marrow-isolated adult multilineage-inducible (MIAMI) cells protect against peripheral vascular ischemia in a mouse model. Cytotherapy. 2011;13(2):179–92.

    Article  CAS  PubMed  Google Scholar 

  112. Grau-Monge C, Delcroix GJ, Bonnin-Marquez A, Valdes M, Awadallah EL, Quevedo DF, Armour MR, Montero RB, Schiller PC, Andreopoulos FM, et al. Marrow-isolated adult multilineage inducible cells embedded within a biologically-inspired construct promote recovery in a mouse model of peripheral vascular disease. Biomed Mater. 2017;12(1): 015024.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Kucia M, Reca R, Campbell FR, Zuba-Surma E, Majka M, Ratajczak J, Ratajczak MZ. A population of very small embryonic-like (VSEL) CXCR4(+)SSEA-1(+)Oct-4+ stem cells identified in adult bone marrow. Leukemia. 2006;20(5):857–69.

    Article  CAS  PubMed  Google Scholar 

  114. Guerin CL, Loyer X, Vilar J, Cras A, Mirault T, Gaussem P, Silvestre JS, Smadja DM. Bone-marrow-derived very small embryonic-like stem cells in patients with critical leg ischaemia: evidence of vasculogenic potential. Thromb Haemost. 2015;113(5):1084–94.

    Article  PubMed  Google Scholar 

  115. Guenther R, Dreschers S, Maassen J, Reibert D, Skazik-Voogt C, Gutermuth A. The Treasury of Wharton’s Jelly. Stem Cell Rev Rep. 2022;18(5):1627–38.

    Article  CAS  PubMed  Google Scholar 

  116. Ratajczak MZ, Ratajczak J, Kucia M. Very Small Embryonic-Like Stem Cells (VSELs). Circ Res. 2019;124(2):208–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Kuroda Y, Kitada M, Wakao S, Nishikawa K, Tanimura Y, Makinoshima H, Goda M, Akashi H, Inutsuka A, Niwa A, et al. Unique multipotent cells in adult human mesenchymal cell populations. Proc Natl Acad Sci U S A. 2010;107(19):8639–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Hosoyama K, Wakao S, Kushida Y, Ogura F, Maeda K, Adachi O, Kawamoto S, Dezawa M, Saiki Y. Intravenously injected human multilineage-differentiating stress-enduring cells selectively engraft into mouse aortic aneurysms and attenuate dilatation by differentiating into multiple cell types. J Thorac Cardiovasc Surg. 2018;155(6):2301-13.e4.

    Article  PubMed  Google Scholar 

  119. Yamada Y, Minatoguchi S, Kanamori H, Mikami A, Okura H, Dezawa M, Minatoguchi S. Stem cell therapy for acute myocardial infarction—focusing on the comparison between Muse cells and mesenchymal stem cells. J Cardiol. 2022;80(1):80–7.

    Article  PubMed  Google Scholar 

  120. Li H, Wei J, Liu X, Zhang P, Lin J. Muse cells: ushering in a new era of stem cell-based therapy for stroke. Stem Cell Res Therapy. 2022; 13(1).

  121. Marchionni C, Bonsi L, Alviano F, Lanzoni G, Di Tullio A, Costa R, Montanari M, Tazzari PL, Ricci F, Pasquinelli G, et al. Angiogenic potential of human dental pulp stromal (stem) cells. Int J Immunopathol Pharmacol. 2009;22(3):699–706.

    Article  CAS  PubMed  Google Scholar 

  122. Sakai VT, Zhang Z, Dong Z, Neiva KG, Machado MA, Shi S, Santos CF, Nör JE. SHED differentiate into functional odontoblasts and endothelium. J Dent Res. 2010;89(8):791–6.

    Article  CAS  PubMed  Google Scholar 

  123. Gong T, Heng BC, Xu J, Zhu S, Yuan C, Lo EC, Zhang C. Decellularized extracellular matrix of human umbilical vein endothelial cells promotes endothelial differentiation of stem cells from exfoliated deciduous teeth. J Biomed Mater Res A. 2017;105(4):1083–93.

    Article  CAS  PubMed  Google Scholar 

  124. Li J, Zhu Y, Li N, Wu T, Zheng X, Heng BC, Zou D, Xu J. Upregulation of ETV2 expression promotes endothelial differentiation of human dental pulp stem cells. Cell Trans. 2021;30:963689720978739.

    Google Scholar 

  125. 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 Res Ther. 2021;12(1):266.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Trimm E, Red-Horse K. Vascular endothelial cell development and diversity. Nat Rev Cardiol. 2023;20(3):197–210.

    Article  PubMed  Google Scholar 

  127. Madfis N, Lin Z, Kumar A, Douglas SA, Platt MO, Fan Y, McCloskey KE. Co-emergence of specialized endothelial cells from embryonic stem cells. Stem Cells Dev. 2018;27(5):326–35.

    Article  CAS  PubMed  Google Scholar 

  128. Li Z, Hu S, Ghosh Z, Han Z, Wu JC. Functional characterization and expression profiling of human induced pluripotent stem cell- and embryonic stem cell-derived endothelial cells. Stem Cells Dev. 2011;20(10):1701–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Homma K, Sone M, Taura D, Yamahara K, Suzuki Y, Takahashi K, Sonoyama T, Inuzuka M, Fukunaga Y, Tamura N, et al. Sirt1 plays an important role in mediating greater functionality of human ES/iPS-derived vascular endothelial cells. Atherosclerosis. 2010;212(1):42–7.

    Article  CAS  PubMed  Google Scholar 

  130. Zhang X, Jiang X, Jiang S, Cai X, Yu S, Pei G. Schwann cells promote prevascularization and osteogenesis of tissue-engineered bone via bone marrow mesenchymal stem cell-derived endothelial cells. Stem Cell Res Ther. 2021;12(1):382.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Bezenah JR, Kong YP, Putnam AJ. Evaluating the potential of endothelial cells derived from human induced pluripotent stem cells to form microvascular networks in 3D cultures. Sci Rep. 2018;8(1):2671.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Halaidych OV, Freund C, van den Hil F, Salvatori DCF, Riminucci M, Mummery CL, Orlova VV. Inflammatory responses and barrier function of endothelial cells derived from human induced pluripotent stem cells. Stem Cell Reports. 2018;10(5):1642–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Graf T, Stadtfeld M. Heterogeneity of embryonic and adult stem cells. Cell Stem Cell. 2008;3(5):480–3.

    Article  CAS  PubMed  Google Scholar 

  134. Finkenzeller G, Graner S, Kirkpatrick CJ, Fuchs S, Stark GB. Impaired in vivo vasculogenic potential of endothelial progenitor cells in comparison to human umbilical vein endothelial cells in a spheroid-based implantation model. Cell Prolif. 2009;42(4):498–505.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

None.

Funding

This work was supported by the Chongqing Talent Program (Grant No. CQYC20210303360).

Author information

Authors and Affiliations

Authors

Contributions

DD participated in the critical editing of the manuscript and image generation. YZ wrote the initial version of the review. ZHZ and BT designed and revised the full manuscript. All authors read and revised multiple versions of the draft and approved the manuscript for submission.

Corresponding authors

Correspondence to Bo Tang or Zhihui Zhang.

Ethics declarations

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Deng, D., Zhang, Y., Tang, B. et al. Sources and applications of endothelial seed cells: a review. Stem Cell Res Ther 15, 175 (2024). https://doi.org/10.1186/s13287-024-03773-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-024-03773-6

Keywords