- Review
- Open access
- Published:
The mechanisms and applications of endothelial progenitor cell therapy in the treatment of intracranial aneurysm
Journal of Translational Medicine volume 23, Article number: 377 (2025)
Abstract
The pathophysiological mechanism of intracranial aneurysm (IA) involves the dynamic interaction of ECM abnormalities, hemodynamic stress, and inflammatory response. The rupture of intracranial aneurysm will cause serious consequences. Multiple studies have confirmed the important role and potential application of endothelial progenitor cells (EPCs) in vascular repair. This review focuses on the specific mechanism of EPCs in the treatment of intracranial aneurysms, which promote re-endothelialization and angiogenesis through bone marrow mobilization, targeted migration to the site of injury, differentiation into mature endothelial cells, and secretion of angiogenic factors. In addition, EPCs maintain ECM homeostasis by regulating MMP/IMP balance, inhibiting aneurysm wall thinning and structural damage. Based on the vascular repair mechanism of EPCs, new treatment strategies such as “biologically active” spring coils (loaded with EPCs or SDF-1α) and flow diverters(FDs) combined with EPCs therapy have been developed to synergistically promote carotid endothelialization of aneurysms and reduce the risk of recurrence. Future research needs to further validate the long-term efficacy and precise regulatory mechanisms of EPCs in clinical translation, providing new directions for IA treatment.
Background
Intracranial aneurysms(IAs), refer to a localized dilation of the arterial walls within the brain. This abnormality most often occurs at branching points in the circle of Willis [1,2,3]. Epidemiological studies suggest that the prevalence of IAs falls between 0.5% and 3% of the general population [2, 4]. When these aneurysms rupture, they can lead to a wide array of severe and life-threatening clinical complications. These may include subarachnoid hemorrhage (SAH), compression of adjacent brain tissue, dysfunction of cranial nerves, hemiparesis, visual disturbances such as defects in the visual field, seizures, pressure on the brainstem, transient ischemic attacks, and in some cases, cerebral infarction due to distal embolization. These conditions can significantly impair neurological function and are associated with a high risk of mortality [2].
The exact origins of IAs are not yet fully elucidated. While genetic factors are suspected to play a significant role in some individuals, other cases may be influenced by external triggers such as infections or inflammatory responses. The development of IAs involves a multifactorial process where genetic predisposition, mechanical forces from blood flow, and abnormalities in the arterial walls collectively contribute to the condition. Several known risk factors, including chronic hypertension, tobacco use, and mutations in specific genes, can lead to the weakening and thinning of the arterial walls, providing the necessary conditions for aneurysm formation and, in some cases, rupture [5,6,7,8]. In addition, the abnormal functioning of certain vascular cells, such as endothelial progenitor cells (EPCs) and vascular smooth muscle cells (VSMCs), which are essential for maintaining and remodeling the vascular wall, has been suggested to play an important role in the pathogenesis of IAs [9, 10].
EPCs are a subtype of cells that originate from CD34+ hematopoietic stem cells in adults. These cells have the remarkable ability to differentiate into endothelial cells when cultured in vitro, which makes them an essential component in vascular repair and regeneration [11]. EPCs contribute significantly to maintaining vascular health by stabilizing the endothelial layer and replenishing damaged endothelial cells, thus ensuring the proper function of blood vessels [12, 13]. EPCs are integral to maintaining the overall health of the vasculature, supporting the repair of damaged vessels and fostering the formation of new blood vessels. Furthermore, these cells have been proposed as biomarkers for assessing cardiovascular disease risk, given their involvement in vascular repair and regeneration [14, 15]. However, under certain conditions such as aging, smoking, hypertension, and IAs, the number of circulating EPCs has been shown to decline significantly [16].
Numerous studies investigating EPCs have unveiled significant therapeutic targets and novel approaches for the treatment and management of cardiovascular diseases. Similarly, gaining a deeper understanding of the roles EPCs play in cerebrovascular conditions is essential for overcoming the challenges in treating IAs and related disorders. It is believed that a disruption in the homeostatic balance between the vascular endothelium and its repair mechanisms increases the vulnerability of the vascular wall to hemodynamic stress, which can ultimately contribute to the formation of IAs [12, 17]. In individuals with IAs, endothelial cells are the first to be affected following vascular injury, and a noticeable decrease in endothelial cell numbers is observed in IAs [18]. The alterations in both the quantity and function of circulating EPCs suggest that these cells are crucial in the pathogenesis of IAs and its progression [19]. Investigations into the role of endothelial repair in IAs development have led to the following conclusions: (1) A decrease in EPC levels is linked to the development of IAs; (2) EPCs are recruited to the aneurysm wall, where they play a significant role in repairing and remodeling the aneurysm; and (3) EPCs promote endothelialization after interventions such as coiling embolization and the implantation of flow diverter (FD) stents [20,21,22,23].
EPCs serve as a fundamental initiating factor in the re-endothelialization process that follows endovascular treatments aimed at occluding aneurysms [24, 25]. Various targeted interventions that aim to enhance the mobilization of EPCs and increase their presence in the bloodstream have shown promising results in preclinical models of IAs [24]. Recent findings show EPCs hold significant promise as a part of therapeutic treatments for IAs. EPCs are recognized as pivotal factors in the endothelialization process that follows blood flow diversion. The neointimal tissue begins to grow along the surface of the device, progressively extending and eventually forming a barrier that isolates the aneurysm from the arteries that supply it [26]. This endothelial coverage, which forms over time, is essential for achieving sustained occlusion of the aneurysm, as it ensures the long-term sealing and isolation of the lesion from the surrounding blood vessels [27].
At present, a variety of treatment options are available for managing IAs. The choice of treatment is determined by assessing several factors, such as the aneurysm size, shape, location, and the condition of its wall, all of which contribute to evaluating the rupture risk. Based on this evaluation, the most suitable treatment approach is selected. In general, the therapeutic strategies include surgical clipping, interventional treatments, and conservative management. The choice of interventional method is often based on the aneurysm’s specific characteristics, such as its shape and size, which influence the selection of materials and techniques. Options for interventional treatment include simple coil embolization, stent-assisted coil embolization, flow-diverting stents, and devices designed to disrupt the aneurysm internally, such as intratumoral spoilers [28,29,30].
The investigation into interactions between EPCs and IAs at pathological stages will help clarify how these conditions develop along with their progressions providing an advanced foundation for creating effective new treatment methods. This review examines EPC functions in IAs management then evaluates EPCs as therapeutic targets to prevent and treat IAs.
Biological characteristics of EPCs
Endothelial precursor cells known as EPCs functionally serve vascular regeneration because they reside mainly within the specialized domains of bone marrow stem cell niches [11]. The presence of EPCs extends beyond their primary bone marrow locale to umbilical cord blood as well as peripheral circulation and arterial walls but detectives discover their quantities diminish in these further locations [31,32,33]. The EPC population shows great heterogeneity because it contains cells at multiple endothelial differentiation phases accompanied by distinctive phenotypic traits and functional abilities. Research has established a characteristic profile for this population using surface markers CD34, CD133, CD31, VEGFR-2, vWF, CD144, Tie2, CD117, CD62E, and CD45 which serve as standard definitions for EPCs [34,35,36,37,38]. EPCs that exhibit CD133+, CD34+, and VEGFR-2+ marker expression represent less mature developmental stages of these cells as identified in reference [35]. The maturation of EPCs results in a progressive decline of specific surface markers including CD133+. These markers are no longer present in mature endothelial cells (ECs), highlighting the dynamic process through which EPCs differentiate into fully functional endothelial cells [39]. Endothelial progenitor cells (EPCs) can be isolated from diverse sources such as bone marrow, peripheral blood, umbilical cord blood, and adipose tissue. Importantly, EPCs from different origins exhibit distinct functional and phenotypic characteristics, which may significantly impact their therapeutic efficacy: Bone marrow-derived EPCs are enriched in CD34+/VEGFR-2 + populations and demonstrate strong migratory capacity toward ischemic tissues, making them ideal for ischemic vascular repair [40]. Peripheral blood-derived EPCs are mobilized in response to hypoxia or cytokines (e.g., SDF-1α) but show lower proliferative potential compared to BM-EPCs [41]. Umbilical cord blood-derived EPCs exhibit higher clonogenic activity and enhanced paracrine secretion of angiogenic factors (e.g., VEGF, Ang-1), suggesting advantages in neovascularization therapies. These source-dependent variations necessitate careful selection of EPCs tailored to specific clinical applications [42].
EPCs are a distinct group of precursor cells that are involved in endothelial cell regeneration and repair. They can be classified into two categories based on their time in culture: early EPCs (eEPCs) and late EPCs (lEPCs) [43]. This classification reflects the different stages of maturation that these cells undergo. eEPCs typically appear within the first 4 to 7 days of culture, whereas lEPCs generally emerge after 14 to 21 days of culturing [44]. eEPCs share phenotypic and functional similarities with CD14+ cells and are often referred to as CD14+ EPCs. Escleman et al. described these cells as originating from the bone marrow or hematopoietic progenitor cells, with a distinct spindle-shaped morphology [45]. The markers typically associated with eEPCs include CD31, CD133, CD34, VEGFR-2, vWF, CD45 (a hematopoietic-specific antigen), and markers like CD14 and CD115, which are characteristic of monocytes and macrophages [35, 46, 47]. These cells are capable of taking up acetylated low-density lipoprotein and binding to the lectin Ulex europeus agglutinin-1 (UEA-1), which are functional traits commonly attributed to mature endothelial cells (ECs) [48]. The combination of these surface markers illustrates their dual functionality, as they exhibit properties of both endothelial and hematopoietic cells. This dual characteristic suggests that eEPCs may contribute to processes such as vascular repair and may also play a role in inflammation.
Late endothelial progenitor cells (lEPCs), sometimes referred to as outgrowth endothelial cells or CD34+ EPCs, are closely related to the circulating CD34+ hematopoietic stem cells that originate in the bone marrow [49]. Cellular cobblestone-like morphology serves as the primary visual distinction between these cells and eEPCs according to reference [50]. lEPCs lack monocyte and macrophage-associated surface markers CD14 and CD115 whereas eEPCs express these markers [51]. Markers CD34 and CD133 along with VEGFR-2 which EPCs express remain inadequate for identification since other cell types share these same markers [52, 53](Fig. 1). The cytokine levels in eEPCs and lEPCs show major differences between the two types. According to research findings from Yoon et al. that eEPCs showed positive expression for CD14 and CD45 markers but lEPCs did not carry the same markers [38]. The research discovered that lEPCs demonstrate higher KDR and chemokine receptor 1(CXCR-1) levels relative to eEPCs. Examination of the cell markers VE-cadherin along with Fms-related receptor tyrosine kinase (Flt-1), KDR and CD45 reveals different expression profiles between these subpopulations. The secretion of angiogenesis-regulating cytokines like VEGF and IL-8 is stronger in eEPCs than in lEPCs when studied in lab cultures yet both EPCs show equal angiogenesis promotion capabilities during live testing [35]. Moreover, surface markers linked to hematopoiesis, such as WAS and LYN, are expressed at significantly lower levels in lEPCs than in eEPCs [54]. In addition, studies by Zhang et al. have shown that eEPCs exhibit limited proliferative potential and are nearly incapable of being passaged, whereas lEPCs demonstrate significantly higher proliferative capacity and the ability to form capillary-like structures, resembling the behavior of microvascular endothelial cells [55].
Two different types of EPC. Early EPCs Originating from bone marrow or hematopoietic progenitor cells, spindle shaped. Cultivation period: 4–7 days. Markers: CD14⁺/CD115⁺ (monocyte/macrophage characteristics), CD31/CD34/CD133/VEGFR-2/CD45⁺ (dual markers of endothelium and hematopoiesis). Function: Intake of Acetylated low-density lipoprotein and binding to UEA-1; Secreted high levels of VEGF/IL-8; Promotes angiogenesis but weak proliferative ability, making it difficult to propagate. Potential role: vascular repair and inflammation regulation. Late EPCs Source and morphology: Derived from bone marrow CD34 ⁺ hematopoietic stem cells, with a pebble like morphology. Cultivation period: 14–21 days. Markers: CD14 ⁻/CD115 ⁻, high expression of KDR/CXCR-1; Low hematopoietic related markers (WAS/LYN). Function: Strong proliferative ability, capable of forming capillary like structures; The in vitro angiogenic activity is comparable to eEPC. Characteristics: It is closer to mature endothelial cells and exhibits microvascular endothelial behavior
However, in terms of the EPCs endovascular treatment of IAs, the cell surface markers of EPCs might play a more important role in the capture, homing, proliferation and differentiation despite the type or origin. One clinical trial in 2005 showed that G-CSF administered to patients increased CD133+/VEGFR-2+ EPCs and endothelial cell-forming clusters in culture [56]. Similarly, it had been demonstrated that one subpopulation of EPCs of CD34−/CD133+ not only significantly showed higher rates of proliferation (P < 0.005), but also a higher potential of celldifferentiation capacity into other cell types which might be applied to the more capture in situ of EPCs through grafting CD133 antibody to surface coating of blood-contact materials [57, 58].
Mechanisms of EPC in the treatment of IAs
The pathophysiology underlying IA is multifactorial, encompassing three main interconnected mechanisms: abnormalities in the ECM, hemodynamic stress, and inflammatory responses. These factors are not only interconnected but also influence each other in a dynamic and complex manner. The ECM plays a crucial role in preserving the mechanical strength and elasticity of the vessel wall. When defects or alterations occur within the ECM, the structural integrity of the vessel is compromised, which can facilitate the formation of aneurysms. One of the key contributing factors to aneurysm formation is increased wall shear stress, which induces endothelial cell injury, smooth muscle cell degradation, and thinning of the vascular media. Changes in blood flow patterns are central to the pathogenesis of aneurysm formation because they influence the localization of inflammatory cells and regulate endothelial cell responses to localized inflammation. Hemodynamic alterations lead to the recruitment of inflammatory cells and the release of pro-inflammatory cytokines, which in turn amplify the local inflammatory environment and perpetuate damage. The endothelial lining, as the primary sensor of changes in shear stress, plays an essential role in modulating inflammatory processes. An intact and functional endothelial monolayer contributes to vascular homeostasis by secreting anti-thrombotic and anti-inflammatory mediators, thus helping protect the vessel wall from excessive inflammation and thrombus formation [59,60,61,62]. The formation of aneurysms is widely believed to be the result of an imbalance between factors that promote endothelial injury and those that initiate repair processes [63, 64]. Injury to endothelial cells sets off an inflammatory cascade in the vessel wall, leading to the accumulation of inflammatory cells and the secretion of cytokines, both of which exacerbate the damage to the vessel wall. This inflammatory response also triggers the phenotypic transition of VSMCs, further contributing to aneurysm formation. Key components of the inflammatory process, such as adhesion molecules, cytokines, ROS, leukocytes, MMPs, and VSMCs, are central to the progression of this pathological state [65, 66]. Among these, monocytes, particularly macrophages, play a critical role in driving the inflammatory cascade. Macrophages release MMP-2 and MMP-9, enzymes that are involved in the breakdown of ECM components and the remodeling of the vascular wall. These enzymes are instrumental in both the formation and rupture of aneurysms, as they degrade the ECM, weaken the vessel wall, and promote structural changes that facilitate aneurysm progression [17, 67].
EPC promotes vascular endothelial repair and vascular wall stability
The vascular endothelium serves as the first line of defense in shielding the vascular walls from the stresses induced by chemical agents, inflammation, and mechanical forces [60]. The body possesses an innate repair system for addressing endothelial damage or dysfunction, which involves the activation of multiple signaling pathways and the recruitment of various repair cells [68]. Research has shown that EPCs play a crucial role in maintaining the integrity and function of the vascular environment by promoting processes such as neovascularization and the re-endothelialization of injured blood vessels [69]. Under normal, healthy conditions, EPCs are primarily located in the bone marrow (BM) microenvironment, which is characterized by low oxygen levels and a high concentration of stromal cell-derived factor-1 alpha (SDF-1α), while their presence in circulating blood is minimal [70]. However, when inflammation, injury, or hypoxia occur, EPCs are “mobilized” from the BM and enter the bloodstream in response to trigger factors such as chemokines, MMP-9, VEGF, and NO produced by damaged blood vessels [71, 72]. The primary factor that initiates the mobilization of EPCs from the BM to the sites of injury in peripheral blood is the release of angiogenic growth factors during ischemic damage, which includes granulocyte-forming factors, GM-CSF, VEGF, and SDF–1α [73, 74]. In response to injury, damaged tissues and platelets release cytokine SDF-1α, which binds to the CXCR-4 receptor on the surface of EPCs, leading to the activation of MMP-9 in combination with growth factors [75]. This interaction also results in the release of SKITL that binds to c-kit, ultimately creating a proliferative BM microenvironment that supports EPC migration [76, 77]. SDF-1α demonstrates additional mobilizing capability by interacting with factors G-CSF and VEGF as well as E-selectin and activating eNOS-dependent signal transduction pathways to drive CXCR4 + stem and progenitor cells from the body into blood circulation [78]. Studies demonstrated that SDF-1 administration leads to elevated VEGF production within ischemic areas and thus raises circulating EPC levels [79]. Research shows infection produces higher plasma amounts of IFN-γ together with VEGF, G-CSF and SDF-1 while also enhancing signal transmission between Sca-1 and RAS-related C3 botulinum toxin substrate 2 (Rac2) within bone marrow-derived EPCs which results in boosted EPCs expression [80].
SDF-1α plays a significant role in regulating the expression of PSGL-1 on progenitor cells [75]. Research has demonstrated that the levels of PSGL-1 expression following vascular injury are dependent on the concentration of SDF-1α, which modulates the response in a dose-dependent manner. PSGL-1 is a ligand for P-selectin, a molecule found on aggregated platelets in damaged blood vessels [81,82,83]. When endothelial damage occurs, platelet activation leads to the release of P-selectin, which in turn binds to PSGL-1 on EPCs, promoting their aggregation at the injury site. In addition, CD34, a surface marker on EPCs, acts as a ligand for E-selectin. The interaction between E-selectin and CD34 enhances the adhesion of EPCs to the ischemic endothelium. Furthermore, SDF-1α amplifies the migration and repair functions of EPCs by stimulating the expression of E-selectin, thereby fostering EPC recruitment to areas of endothelial damage [84]. Based on this, the above research proved the application potential of SDF-1α in vascular wall repair. Studies that applied SDF-1α-together-coated coils established that autologous mesenchymal stem cells administration provided structural strengthening of the aneurysm sac while decreasing its volume [21].
EPCs play a pivotal role in vascular repair by adhering to sites of tissue injury through integrins. Upon attachment, EPCs contribute to neovascularization and re-endothelialization by transforming into mature ECs and remodeling the extracellular matrix components of the damaged tissue [85, 86]. The paracrine signals derived from EPCs play a key role in creating an angiogenesis-promoting microenvironment that supports both the formation of new blood vessels and the regeneration of the existing vasculature. Upon mobilization, EPCs further attract various angiogenic growth factors such as G-CSF, GM-CSF, HGF, and VEGF [48]. These growth factors activate various intracellular signaling pathways, including those involving phosphatidylinositol 3-kinase(PI3K) and mitogen-activated protein kinase(MAPK) [87]. The downstream effects of these pathways contribute to the creation of a microenvironment that facilitates endothelial cell proliferation and supports tissue repair in the affected area.
EPCs are crucial in maintaining endothelial integrity by producing angiogenic factors that promote the proliferation, survival, and functionality of mature ECs, as well as other neighboring progenitor cells, such as smooth muscle progenitor cells and other vascular progenitors [88]. EPCs also possess the ability to migrate toward areas of injury, where they differentiate into functional endothelial cells, restore the integrity of the endothelial barrier, enhance the stability of the vascular wall, and decrease the likelihood of aneurysm rupture by upregulating the expression of VEGF, SDF-1α, and CXCR4 [89]. Through modulation of MMP-2 and MMP-9 levels against their TIMP inhibitors, EPCs prevent structural tissue damage to elastin and collagen sustaining vascular wall durability. According to studies conducted on rodents the injection of EPCs demonstrates both the reduction of aneurysm wall thinning along with the maintenance of the inner elastic layer and promotion of new extracellular matrix deposition within the vessel wall [90].
In view of the problem that the endothelial coverage rate of intracranial aneurysms after traditional coil embolization is less than 50% and easy to relapse, “bioactive” coils have been developed to improve the long-term stability of the procedure, ongoing research remains dedicated to enhancing the rate of endothelialization at the aneurysmal neck. At 16 weeks post-treatment patients treated with EPCs developed uniform endothelial cell layers along with beneath neointima formations across their aneurysm neck [20]. The current research reveals that EPCs show targeted movement toward coil curls leading to endothelial cell expansion which supports the entire endothelialization mechanism [24]. Different microRNAs have become the focus of additional research for potential impacts on endothelialization rates after coil embolization. Yu et al. discovered that mir-31a-5p microRNA generates enhanced circulation and functional ability for EPCs to migrate and make new endothelial layers [91].
Flow diverter and EPCs synergistically promote endothelialization. Immediately after the flow diversion treatment, the first observable event is the adhesion of clusters of inflammatory cells to the neck of the aneurysm, injury to the vessel’s endothelial layer triggers a cascade of responses, leading to the release of several cytokines, including VEGF, FGF, SDF-1α, NO, CXCR-8, and Angiopoietin-1 [92]. These signaling molecules contribute to the activation and proliferation of local endothelial cells as well as SMCs, which play essential roles in the healing process of the injured tissue [93]. Additionally, these cytokines promote the migration of EPCs toward the site of vascular damage [94](Fig. 2).
Flow diverter and EPCs synergistically promote endothelialization. After stent implantation, inflammation of the vascular wall is triggered firstly, attracting inflammatory cells and a series of inflammatory factors are released including VEGF, FGF, SDF-1α, NO, CXCR-8, and Angiopoietin-1 these cytokines promote the proliferation and migration of EPCs toward the site of vascular damage. After the mobilization, the surface markers PSGL-1 and CD34 are combined with P-selectin and E-selectin accordingly. Then the EPC differentiate into endothelial cells along the scaffold, and a new endothelial layer is formed eventually. The damaged endothelial cells also release some cytokines like VEGF, SDF-1α, GM-CSF, which can activate MMP-9, then trigger the combination of SKITL and c-Kit that enhance the proliferation and mobilization of EPCs
Angiopoietin-1 is an important pro-angiogenic factor, known for its pivotal function in stimulating vascular remodeling. It achieves this by interacting with the Tie-2 receptor, which is expressed on the surface of endothelial cells [95]. The role of this signaling pathway becomes particularly crucial within the bone marrow, where it helps to maintain hematopoietic stem cells in an undifferentiated state. After ischemic injury, the expression of Angiopoietin-1 increases significantly in brain endothelial cells, suggesting its vital involvement in vascular repair mechanisms [96, 97].
Endothelialization, a process that takes time, is primarily driven by endothelial cells originating from the adjacent parent artery, making it a gradual event [22]. Simultaneously, the structural elements of the FD itself, namely the struts, act as a supportive scaffold, aiding the migration of endothelial cells from both the proximal and distal sections of the parent artery [98]. Endothelialization occurs in two distinct phases: the first is a rapid, early phase characterized by the widespread migration of cells from the surrounding tissue, followed by a slower, later phase where the device’s structure plays a supportive role. During this latter phase, EPCs migrate to the injury site, aided by the scaffolding provided by the stent. EPCs originating from the bone marrow are essential for the neointima formation and re-endothelialization processes that occur after the aneurysm is treated with elastase-induced blood flow diversion. These EPCs can differentiate into various cell types depending on the progression of neointima formation, demonstrating their plasticity and adaptability in response to the tissue environment [99]. In preclinical animal models, the process of endothelialization following the implantation of a FD typically spans a period of 4 to 8 weeks. However, early changes can be observed as soon as the first day after the deployment of the stent, with the appearance of patchy distributions of CD31+ endothelial cells along the metal struts of the device [22]. This initial response is a critical part of the vascular healing process. Further research into endothelialization in blood flow guidance devices has been conducted using rabbit aneurysm models, where the migration and behavior of EPCs have been closely observed. Utilizing immunolabeled scanning electron microscopy with CD34+ markers, it was found that by the 60th day after implantation, up to 87–90% of the surface area of the device was covered by new tissue growth [100].The mechanisms of EPCs in the treatment of intracranial aneurysms are showed in the Fig. 2.
Based on the basic idea of promoting EPCs migration and strengthening endothelialization, many studies have been carried out on FD stent coating. Modern coating advances for FD stents aim to reduce thrombotic risk while actively driving neointimal growth that stabilizes long-term vessel repair. The current biomedical research focuses on developing new stents covered with biofactors that produce two biological actions which serve anticoagulation and endothelialization. The field of cerebrovascular interventions has gained a promising new treatment strategy which uses stents covered with anti-CD34 antibodies. The current treatment emerged from previous findings which showed these coatings enhance EPC migration to damaged vascular sites and improve recovery for patients with aneurysms and vascular problems [101]. FD stents now benefit from a significant development through the application of CD31 surface coatings. The transmembrane glycoprotein CD31 recognized as PECAM-1 facilitates both adhesion and movement within endothelial cells. Clinical studies verify that FD stents perform better when CD31 coatings help them bond with the vascular wall because of the importance of device integration for treatment success. When applied the coating becomes dual-purpose by increasing both endothelial cell adhesion to the surface and reducing inflammatory responses that create favorable conditions for healing. The histological examination of the arterial wall created using CD31-coated FD stents at the aneurysm neck shows evidence of neovascularization structure development. The newly formed endothelial structure appears like arterial middle layers through its distinct thick collagen composition positioned adjacent to a strong smooth muscle cell arrangement [102].
Dual-action stents aim to better aneurysm treatment outcomes by delivering improved occlusion while reducing the complication risks of endovascular interventions. Construction of biofactor-coated scaffolds has been accomplished with nickel-titanium alloy sheets. The scaffold sheets contain both VEGF and anti-CD34 antibodies which induce endothelial cell growth while supporting EPC recognition and adhesion. Scientists have proven that nickel-titanium alloy sheets support bioactive scaffolds to carry VEGF and anti-CD34 antibodies which function as principal drivers of the healing mechanism. The experimental results demonstrate these scaffolds possess superior blood compatibility in research tests. Though these materials support both the growth of human umbilical vein endothelial cells (HUVECs) they help to bind the EPCs effectively. These results reveal biofactor-coated scaffolds show promise as essential tools for better aneurysm intervention results and safe endovascular practice which promises substantial clinical application opportunities [103]. In the year 2021 Pipeline Flex Embolization Device (PED) shield technology within the Pipeline Flex system secured FDA marketing approval as the third-generation blood flow guidance device. The first design purpose of this device focused on thrombosis reduction through choline phosphate coating. Scientific investigations show that PED shield enhances early endothelial cell development during pig aneurysm procedure testing into practice [104]. Clinical evaluations of the PED shield revealed safe performance data because it achieved occlusion rates between 81% and 92% throughout a one-year period according to studies [105,106,107].
Recent advancements in bioengineering have increasingly focused on developing biodegradable scaffolds with integrated cell-therapeutic functions hat hold regenerative potential for vascular applications. A variety of biodegradable scaffold designs are being explored as promising options for the treatment of IA, particularly when combined with EPC delivery to address both structural and biological repair. Among the most commonly utilized materials for biodegradable scaffolds are PLA and PLGA, both of which offer significant advantages in terms of biocompatibility and controlled degradation [92]. Upon implantation of a blood flow-guided stent, the release of cytokines stimulates endothelial and smooth muscle cell activity, which in turn promotes the mobilization of EPCs to the site of vascular injury. Moreover, scaffolds that are coated with antibodies capable of capturing EPCs and facilitating endothelialization present an exciting potential therapeutic approach in the future [92]. Emerging strategies such as 3D-bioprinted hybrid scaffolds pre-seeded with EPCs demonstrate a promising treatment for enhancing bone defect repair which is hopeful for the treatment of IAs [108].And hydrogel surfaces have been improved to enhance the attachment and adhesion of EPCs [109].
Nishi and colleagues were pioneers in developing the first fully bioabsorbable FD made from 48 braided poly-L-lactic acid fibers [110]. Further investigation by Jamshidi et al. using PLA revealed that in some cases, stent vessel wall non-adherence led to local thrombosis, raising concerns about the safety of these materials [111].
During vascular healing processes, EPCs provide essential endothelial cell support so that through their angiogenic factor secretion including VEGF and FGF and the cytokine IL-10, they promote proper vessel regeneration. New blood vessel development alongside existing vasculature regeneration results from these environmental support constructs. EPCs help generate a protective endothelial layer at treated aneurysm sites that prevents platelet aggregation while reducing thrombosis rates resulting in decreased recurrences. NO and PGI2, both secreted by EPCs, play a key role in providing antithrombotic protection by inhibiting platelet activation and aggregation [112, 113]. Thrombosis often complicates interventional treatments, particularly after embolization and blood flow diversion procedures, contributing to treatment failure. EPCs can mitigate this risk by contributing to the formation of a complete endothelial layer in the aneurysm neck, which decreases platelet adhesion and, as a result, reduces the risk of clot formation. Furthermore, EPCs are instrumental in the repair of vascular endothelium, restoring smooth vascular surfaces, reducing the occurrence of thrombosis in areas of endothelial injury, and minimizing complications that can arise after interventional procedures [114, 115]. It has been demonstrated that the CD146 Ab-armed nanofilamentous stent could show great performance in the reduction of thrombosis and restenosis through re-endothelialization due to highly efficient late EPCs cell capture [116].In a clinical trial which aimed to evaluate whether quantitation of peripheral blood endothelial progenitor cells (EPCs) could improve prediction of unprovoked venous thrombosis(VTE) recurrence risk showed that levels of EPCs were lower in patients who developed VTE recurrence [117].
In individuals suffering from IA, the concentration of EPCs in circulation is typically reduced. In addition, their migration, adhesion, and senescence capabilities are often enhanced, which may contribute to disease progression [19, 118]. Numerous studies have utilized various clinical pharmacological agents to stimulate the mobilization of EPCs and direct their recruitment to sites of aneurysm formation. For instance, preclinical animal models have shown that statins, including rosuvastatin and atorvastatin, effectively promote the movement of EPCs from the bone marrow. This enhanced EPC mobilization facilitates endothelialization at the aneurysm neck, which helps reduce aneurysm degeneration and contributes to overall vascular healing [119, 120]. Further research has demonstrated that autologous blood transfusions involving EPCs result in these cells preferentially migrating to the neointima of damaged blood vessels [121]. Additionally, elevated erythropoietin levels have been shown to stimulate the migration of EPCs to injured areas [16], while sitagliptin has been found to support vascular repair by enhancing EPC migration and aiding the healing process of damaged vasculature [122].
Regulate local inflammatory response
An increased local inflammatory reaction along with oxidative stress occurs during aneurysm development. Macrophages and T cells move into injured blood vessels while secreting inflammatory substances including TNF-α and IL-1β together with MMPs. The production of ROS undergoes simultaneous elevation. The combined factors lead to significant vascular wall deterioration and enlarged aneurysm expansion over time [123]. EPCs control inflammatory progression by blocking macrophage activation together with their migratory activity. The reduction of pro-inflammatory factors achieves the slowing of local inflammation while simultaneously promoting vascular stabilization. Endothelial Progenitor Cells release anti-inflammatory cytokines like IL-10 and TIMP-1 to control local immune response and reduce inflammation-related vascular damage after vessel wall rupture events [124, 125]. Research identified that EPCs block MMP activity, including that of MMP-2 and MMP-9 which results in decreased degradation of the aneurysm wall matrix. Such treatment causes aneurysm walls to heal and slows down both their outward growth and deteriorating progression. EPC intervention creates favorable conditions that allow therapeutic approaches to function more effectively in retreating aneurysms [90]. New studies demonstrate how exosomes produced by EPCs demonstrate promising therapeutic potential. According to Ma et al.‘s study endothelial cells generate dangerous levels of ROS after hypoxia/reoxygenation(H/R) trauma leading to vascular malfunction. Under H/R injury endothelial cells undergo increased apoptosis which EPC-derived extracellular vesicles help to reduce by also protecting against excessive ROS production thereby demonstrating EPC potential for vascular regenerative therapy [125].
The treatment of IAs through interventional methods is frequently combined with drug therapies to enhance the process of endothelialization at the aneurysmal neck. Among these drugs, aspirin is widely acknowledged for its antiplatelet and anti-inflammatory effects. It has been demonstrated in animal models, particularly rats, to notably reduce degeneration of the aneurysmal wall. This protective effect is attributed to the increased mobilization of EPCs and a reduction in chronic vascular inflammation mediated by macrophages [126]. In the same aneurysmal tissues, RT-PCR analysis has shown a reduction in the expression of various inflammatory markers, including NF-κB, MCP-1, and VCAM-1, in mice treated with cilostazol when compared to those in the control group [127]. Additionally, a recent study by Suzuki et al. [128] reinforced the beneficial impact of Cilostazol on the rupture rates of induced IAs in mice. While the formation rate of aneurysms between the Cilostazol-treated group and the control group did not present significant statistical differences, a noteworthy reduction in the rupture rate was observed in the Cilostazol group. In addition to these findings, statins and aspirin have been shown to not only promote the proliferation of EPCs but also inhibit inflammatory responses in the aneurysmal wall. These pharmacological agents reduce the expression of several key inflammatory factors, including iNOS, MMP-2, MMP-9, VEGF, NF-κB, and MCP-1. Previous studies have highlighted the role of sustained hemodynamic shear stress, which leads to endothelial dysfunction at vascular bifurcations, triggering inflammatory responses. This inflammatory activation is followed by the invasion of immune cells such as macrophages, T lymphocytes, and B lymphocytes into the aneurysmal tissue, which leads to further damage of the vascular wall [12, 129]. Therefore, these dual-action drugs, which stimulate EPC mobilization while simultaneously reducing inflammation, may be more effective in stabilizing the aneurysmal wall and preventing rupture compared to therapies that solely focus on enhancing EPC numbers.
EPCs can be relatively easily isolated from tissues such as bone marrow or blood in research settings. However, their clinical translation necessitates rigorous manufacturing processes to ensure safety and efficacy: EPCs must be purified from heterogeneous cell populations and expanded under standardized culture conditions. And cells require extensive testing for viability, sterility, genetic stability, and functional potency (e.g., differentiation capacity, cytokine secretion). Clinical-grade EPCs must be produced in Good Manufacturing Practice-certified facilities, adhering to strict protocols for traceability and contamination control. Skilled technicians and quality assurance teams are essential to manage these complex workflows. These challenges underscore the gap between laboratory-scale EPC isolation and scalable clinical production. But multiple studies have confirmed the separation method of EPC and its safe application in clinical practice [130].
For the application of stem cells, preclinical testing using animal models is particularly important because stem cells can function through multiple mechanisms, and it is difficult to predict the situation of stem cells in animals under cell culture conditions. The integration of physiological functions and permanent tissue reconstruction is the goal of stem cell therapy for many diseases. Animal models will help detect potential side effects of transplanted cell products. Animal model testing is particularly necessary when cells undergo extensive in vitro processing and/or when cell products come from pluripotent stem cells. It must be acknowledged that preclinical testing, including animal model studies, can provide limited evidence on the response of human cell transplantation in humans, as cell behavior also depends on the cell environment and the recipient’s immune response. The independent peer review of preclinical data must take into account this uncertainty. Only under the condition that preclinical data has sufficient persuasiveness, can clinical trials be cautiously and progressively conducted on appropriate patients under strict and independent scientific and ethical supervision [131].
According to ISSCR Guidelines for the Clinical Translation of Stem Cells: chemical or recombinant protein products can be produced with high purity, and the cells produced or collected and prepared from different anatomical locations or individuals have outstanding biological diversity issues. In allogeneic treatment, identifying a single source of main cells can reduce this variability. However, the cell sources for autologous therapy are relatively limited, making it impossible to conduct extensive quality testing. Given that researchers generally lack experience in production, cultivation, and use, the definition of the properties and potential of stem cells and their derivatives still needs to be determined in future research processes [132].
Summary
In the field of treatment of intracranial aneurysms, endothelial progenitor cells (EPCs) show unique potential by promoting vascular endothelial repair, inhibiting inflammatory response and regulating vascular remodeling, which provides a new strategy for clinical intervention. However, at present, the application of EPCs still faces many challenges: the limited source of autologous EPCs and the insufficient expansion efficiency in vitro, the low survival rate of transplanted cells and the functional heterogeneity limit its large-scale application; In addition, the dynamic regulation mechanism of EPCs in the complex microenvironment of aneurysms has not been fully clarified, and the long-term efficacy and safety still need more clinical data to support. Future research needs to focus on the unification of separation and identification standards of EPCs, and the development of targeted delivery system based on biomaterials. Only through interdisciplinary collaboration and translational medicine innovation can we break through the existing bottleneck and promote the efficient transformation of EPC therapy from experimental platform to clinical practice.
Data availability
Not applicable.
Abbreviations
- BM:
-
Bone Marrow
- CXCR-1:
-
Chemokine Receptor 1
- ECs:
-
Endothelial Cells
- EPCs:
-
Endothelial Progenitor Cells
- FD:
-
Flow Diverter
- Flt-1:
-
Fms-related receptor tyrosine kinase
- H/R:
-
Hypoxia/Reoxygenation
- HUVECs:
-
Human Umbilical Vein Endothelial Cells
- IA:
-
Intracranial Aneurysm
- PECAM-1:
-
Platelet Endothelial Cell Adhesion Molecule-1
- PED:
-
Pipeline Flex Embolization Device
- PI3K/MAPK:
-
Phosphatidylinositol 3-kinase/ Mitogen-Activated Protein Kinase
- PGI2:
-
Prostaglandin-I-2
- PSGL-1:
-
P-Selectin Glycoprotein Ligand-1
- Rac2:
-
RAS-related C3 botulinum toxin substrate 2
- SAH:
-
Subarachnoid Hemorrhage
- SDF-1α:
-
Stromal cell-Derived Factor-1 alpha
- UEA-1:
-
Ulex Europeus Agglutinin-1
- VSMCs:
-
Vascular Smooth Muscle Cells
- VTE:
-
Venous Thrombosis
References
Vlak MH, Algra A, Brandenburg R, Rinkel GJ. Prevalence of unruptured intracranial aneurysms, with emphasis on sex, age, comorbidity, country, and time period: a systematic review and meta-analysis. Lancet Neurol Jul. 2011;10(7):626–36.
Brown RD Jr., Broderick JP. Unruptured intracranial aneurysms: epidemiology, natural history, management options, and Familial screening. Lancet Neurol Apr. 2014;13(4):393–404.
Claassen J, Park S. Spontaneous subarachnoid haemorrhage. Lancet Sep. 2022;10(10355):846–62.
Tawk RG, Hasan TF, D’Souza CE, Peel JB, Freeman WD. Jul. Diagnosis and Treatment of Unruptured Intracranial Aneurysms and Aneurysmal Subarachnoid Hemorrhage. Mayo Clin Proc. 2021;96(7):1970–2000.
Brisman JL, Song JK, Newell DW. Cerebral aneurysms. N Engl J Med Aug. 2006;31(9):928–39.
Etminan N, Rinkel GJ. Unruptured intracranial aneurysms: development, rupture and preventive management. Nat Rev Neurol Dec. 2016;12(12):699–713.
Frösen J, Cebral J, Robertson AM, Aoki T. Flow-induced, inflammation-mediated arterial wall remodeling in the formation and progression of intracranial aneurysms. Neurosurg Focus Jul. 2019;1(1):E21.
Toader C, Eva L, Bratu BG et al. Intracranial aneurysms and genetics: an extensive overview of genomic variations, underlying molecular dynamics, inflammatory indicators, and Forward-Looking insights. Brain Sci Oct 12 2023;13(10).
Lee S, Kim IK, Ahn JS, et al. Deficiency of endothelium-specific transcription factor Sox17 induces intracranial aneurysm. Circulation Mar. 2015;17(11):995–1005.
Wei H, Mao Q, Liu L, et al. Changes and function of Circulating endothelial progenitor cells in patients with cerebral aneurysm. J Neurosci Res Nov. 2011;89(11):1822–8.
Asahara T, Murohara T, Sullivan A, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Sci Feb. 1997;14(5302):964–7.
Deanfield JE, Halcox JP, Rabelink TJ. Endothelial function and dysfunction: testing and clinical relevance. Circulation Mar. 2007;13(10):1285–95.
Gill M, Dias S, Hattori K, et al. Vascular trauma induces rapid but transient mobilization of VEGFR2(+)AC133(+) endothelial precursor cells. Circ Res Feb. 2001;2(2):167–74.
Kou F, Zhu C, Wan H, et al. Endothelial progenitor cells as the target for cardiovascular disease prediction, personalized prevention, and treatments: progressing beyond the state-of-the-art. Epma J Dec. 2020;11(4):629–43.
Zhang K, Liu T, Li JA, Chen JY, Wang J, Huang N. Surface modification of implanted cardiovascular metal stents: from antithrombosis and antirestenosis to endothelialization. J Biomed Mater Res Feb. 2014;102(2):588–609.
Xu Y, Tian Y, Wei HJ, et al. Erythropoietin increases Circulating endothelial progenitor cells and reduces the formation and progression of cerebral aneurysm in rats. Neurosci May. 2011;5:181:292–9.
Aoki T, Kataoka H, Morimoto M, Nozaki K, Hashimoto N. Macrophage-derived matrix metalloproteinase-2 and– 9 promote the progression of cerebral aneurysms in rats. Stroke Jan. 2007;38(1):162–9.
Jamous MA, Nagahiro S, Kitazato KT, et al. Endothelial injury and inflammatory response induced by hemodynamic changes preceding intracranial aneurysm formation: experimental study in rats. J Neurosurg Aug. 2007;107(2):405–11.
Liang C, Feng H, Deng BQ, et al. Decreased levels and function of Circulating endothelial progenitor cells in unruptured intracranial saccular aneurysm patients. Neurol Sci Jan. 2014;35(1):23–8.
Aronson JP, Mitha AP, Hoh BL, et al. A novel tissue engineering approach using an endothelial progenitor cell-seeded biopolymer to treat intracranial saccular aneurysms. J Neurosurg Sep. 2012;117(3):546–54.
Gao Y, Lu Z, Chen C, et al. Mesenchymal stem cells and endothelial progenitor cells accelerate intra-aneurysmal tissue organization after treatment with SDF-1α-coated coils. Neurol Res Apr. 2016;38(4):333–41.
Kadirvel R, Ding YH, Dai D, Rezek I, Lewis DA, Kallmes DF. Cellular mechanisms of aneurysm occlusion after treatment with a flow diverter. Radiol Feb. 2014;270(2):394–9.
Li Z, Zhao R, Fang X, Huang Q, Liu J. Recombinant human SDF-1α administration accelerates aneurysm neck reendothelialization in rabbit saccular aneurysm after flow diverter treatment. Acta Biochim Biophys Sin (Shanghai). Mar 1. 2017;49(3):246–253.
Liu P, An Q, Chen X, Huang J, Yang GY, Zhu W. Rosuvastatin for enhancement of aneurysm neck endothelialization after coil embolization: promotion of endothelial progenitor cells in a rodent model. J Neurosurg May. 2016;124(5):1265–74.
Li J, Ma Y, Miao XH, Guo JD, Li DW. Neovascularization and tissue regeneration by endothelial progenitor cells in ischemic stroke. Neurol Sci Sep. 2021;42(9):3585–93.
Sadasivan C, Cesar L, Seong J, et al. An original flow diversion device for the treatment of intracranial aneurysms: evaluation in the rabbit elastase-induced model. Stroke Mar. 2009;40(3):952–8.
Dmytriw AA, Phan K, Moore JM, Pereira VM, Krings T, Thomas AJ. On flow diversion: the changing landscape of intracerebral aneurysm management. AJNR Am J Neuroradiol Apr. 2019;40(4):591–600.
Pontes FGB, da Silva EM, Baptista-Silva JC, Vasconcelos V. Treatments for unruptured intracranial aneurysms. Cochrane Database Syst Rev May. 2021;10(5):Cd013312.
Gaub M, Murtha G, Lafuente M et al. Flow diversion for endovascular treatment of intracranial aneurysms: past, present, and future directions. J Clin Med Jul 16 2024;13(14).
Sahnoun M, Soize S, Manceau PF, Gelmini C, Pierot L. Intracranial aneurysm treatment with WEB and adjunctive stent: preliminary evaluation in a single-center series. J Neurointerv Surg Feb. 2022;14(2):164–8.
Doyle B, Caplice N. A new source of endothelial progenitor cells–vascular biology redefined? Trends Biotechnol Sep. 2005;23(9):444–6.
Finney MR, Greco NJ, Haynesworth SE, et al. Direct comparison of umbilical cord blood versus bone marrow-derived endothelial precursor cells in mediating neovascularization in response to vascular ischemia. Biol Blood Marrow Transpl May. 2006;12(5):585–93.
Pacilli A, Pasquinelli G. Vascular wall resident progenitor cells: a review. Exp Cell Res Apr. 2009;1(6):901–14.
Rouhl RP, van Oostenbrugge RJ, Damoiseaux J, Tervaert JW, Lodder J. Endothelial progenitor cell research in stroke: a potential shift in pathophysiological and therapeutical concepts. Stroke Jul. 2008;39(7):2158–65.
Hur J, Yoon CH, Kim HS, et al. Characterization of two types of endothelial progenitor cells and their different contributions to neovasculogenesis. Arterioscler Thromb Vasc Biol Feb. 2004;24(2):288–93.
Zhao YH, Yuan B, Chen J, et al. Endothelial progenitor cells: therapeutic perspective for ischemic stroke. CNS Neurosci Ther Feb. 2013;19(2):67–75.
Cheng CC, Chang SJ, Chueh YN, et al. Distinct angiogenesis roles and surface markers of early and late endothelial progenitor cells revealed by functional group analyses. BMC Genomics Mar. 2013;15:14:182.
Yoon CH, Hur J, Park KW, et al. Synergistic neovascularization by mixed transplantation of early endothelial progenitor cells and late outgrowth endothelial cells: the role of angiogenic cytokines and matrix metalloproteinases. Circulation Sep. 2005;13(11):1618–27.
Gehling UM, Ergün S, Schumacher U, et al. In vitro differentiation of endothelial cells from AC133-positive progenitor cells. Blood May. 2000;15(10):3106–12.
Chen C, Dai P, Nan L, et al. Isolation and characterization of endothelial progenitor cells from canine bone marrow. Biotech Histochem Feb. 2021;96(2):85–93.
Tanaka R, Masuda H, Fujimura S, et al. Quality-Quantity control culture enhances vasculogenesis and wound healing efficacy of human diabetic peripheral blood CD34 + Cells. Stem Cells Transl Med. May 2018;7(5):428–38.
Kim J, Kim M, Jeong Y, et al. BMP9 induces cord Blood-Derived endothelial progenitor cell differentiation and ischemic neovascularization via ALK1. Arterioscler Thromb Vasc Biol Sep. 2015;35(9):2020–31.
Patel J, Donovan P, Khosrotehrani K. Concise review: functional definition of endothelial progenitor cells: A molecular perspective. Stem Cells Transl Med Oct. 2016;5(10):1302–6.
Yang HM, Hur J, Yoon CH, et al. A novel subset of T cells (angiogenic T cells) facilitates EPC differentiation and has clinical relevance. Am J Cardiol Oct. 2007;100(8A):L80–80.
Rehman J, Li JL, Orschell CM, March KL. Peripheral blood endothelial progenitor cells are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation Mar. 2003;107(8):1164–9.
Sieveking DP, Buckle A, Celermajer DS, Ng MK. Strikingly different angiogenic properties of endothelial progenitor cell subpopulations: insights from a novel human angiogenesis assay. J Am Coll Cardiol. Feb 2008;12(6):660–8.
Niederseer D, Steidle-Kloc E, Mayr M, et al. Effects of a 12-week alpine skiing intervention on endothelial progenitor cells, peripheral arterial tone and endothelial biomarkers in the elderly. Int J Cardiol. Jul 2016;1:214:343–7.
Rehman J, Li J, Orschell CM, March KL. Peripheral blood endothelial progenitor cells are derived from monocyte/macrophages and secrete angiogenic growth factors. Circulation Mar. 2003;4(8):1164–9.
Zhang K, Liu T, Li JA, Chen JY, Wang J, Huang N. Surface modification of implanted cardiovascular metal stents: from antithrombosis and antirestenosis to endothelialization. J Biomedical Mater Res Part Feb. 2014;102(2):588–609.
Ingram DA, Mead LE, Tanaka H, et al. Identification of a novel hierarchy of endothelial progenitor cells using human peripheral and umbilical cord blood. Blood Nov. 2004;104(9):2752–60.
Yoder MC, Mead LE, Prater D, et al. Redefining endothelial progenitor cells via clonal analysis and hematopoietic stem/progenitor cell principals. Blood Mar. 2007;1(5):1801–9.
Madonna R, De Caterina R. Circulating endothelial progenitor cells: do they live up to their name? Vascular Pharmacology. Apr-Jun. 2015;67–69:2–5.
Peichev M, Naiyer AJ, Pereira D, et al. Expression of VEGFR-2 and AC133 by Circulating human CD34(+) cells identifies a population of functional endothelial precursors. Blood Feb. 2000;1(3):952–8.
Kaushik K, Das A. Endothelial progenitor cell therapy for chronic wound tissue regeneration. Cytotherapy Nov. 2019;21(11):1137–50.
Wils J, Favre J, Bellien J. Modulating putative endothelial progenitor cells for the treatment of endothelial dysfunction and cardiovascular complications in diabetes. Pharmacol Ther Feb. 2017;170:98–115.
Powell TM, Paul JD, Hill JM, et al. Granulocyte colony-stimulating factor mobilizes functional endothelial progenitor cells in patients with coronary artery disease. Arterioscler Thromb Vasc Biol Feb. 2005;25(2):296–301.
Bachelier K, Bergholz C, Friedrich EB. Differentiation potential and functional properties of a CD34–CD133 + subpopulation of endothelial progenitor cells. Mol Med Rep Jan. 2020;21(1):501–7.
Duan Y, Yu S, Xu P, et al. Co-immobilization of CD133 antibodies, vascular endothelial growth factors, and REDV peptide promotes capture, proliferation, and differentiation of endothelial progenitor cells. Acta Biomater Sep. 2019;15:96:137–48.
Penn DL, Komotar RJ, Sander Connolly E. Hemodynamic mechanisms underlying cerebral aneurysm pathogenesis. J Clin Neurosci Nov. 2011;18(11):1435–8.
Aoki T, Nishimura M, Matsuoka T, et al. PGE(2) -EP(2) signalling in endothelium is activated by haemodynamic stress and induces cerebral aneurysm through an amplifying loop via NF-κB. Br J Pharmacol Jul. 2011;163(6):1237–49.
Aoki T, Nishimura M, Kataoka H, Ishibashi R, Nozaki K, Miyamoto S. Complementary Inhibition of cerebral aneurysm formation by eNOS and nNOS. Lab Invest Apr. 2011;91(4):619–26.
Tzima E, Irani-Tehrani M, Kiosses WB, et al. A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nat Sep. 2005;15(7057):426–31.
Soldozy S, Norat P, Elsarrag M, et al. The biophysical role of hemodynamics in the pathogenesis of cerebral aneurysm formation and rupture. Neurosurg Focus Jul. 2019;1(1):E11.
Xu Z, Rui YN, Hagan JP, Kim DH. Intracranial aneurysms: pathology, genetics, and molecular mechanisms. Neuromolecular Med Dec. 2019;21(4):325–43.
Chalouhi N, Ali MS, Starke RM, et al. Cigarette smoke and inflammation: role in cerebral aneurysm formation and rupture. Mediators Inflamm. 2012;2012:271582.
Chalouhi N, Points L, Pierce GL, Ballas Z, Jabbour P, Hasan D. Localized increase of chemokines in the lumen of human cerebral aneurysms. Stroke Sep. 2013;44(9):2594–7.
Chalouhi N, Ali MS, Jabbour PM, et al. Biology of intracranial aneurysms: role of inflammation. J Cereb Blood Flow Metab Sep. 2012;32(9):1659–76.
Evans CE, Iruela-Arispe ML, Zhao YY. Mechanisms of endothelial regeneration and vascular repair and their application to regenerative medicine. Am J Pathol Jan. 2021;191(1):52–65.
Sobhan PK, Seervi M, Joseph J, et al. Immortalized functional endothelial progenitor cell lines from umbilical cord blood for vascular tissue engineering. Tissue Eng Part C Methods Nov. 2012;18(11):890–902.
Balaji S, King A, Crombleholme TM, Keswani SG. The role of endothelial progenitor cells in postnatal vasculogenesis: implications for therapeutic neovascularization and wound healing. Adv Wound Care (New Rochelle). Jul 2013;2(6):283–95.
Velazquez OC. Angiogenesis and vasculogenesis: inducing the growth of new blood vessels and wound healing by stimulation of bone marrow-derived progenitor cell mobilization and homing. J Vasc Surg Jun. 2007;45:A39–47. Suppl A(Suppl A).
Tilling L, Chowienczyk P, Clapp B. Progenitors in motion: mechanisms of mobilization of endothelial progenitor cells. Br J Clin Pharmacol Oct. 2009;68(4):484–92.
Urbich C, Heeschen C, Aicher A, Dernbach E, Zeiher AM, Dimmeler S. Relevance of monocytic features for neovascularization capacity of Circulating endothelial progenitor cells. Circulation Nov. 2003;18(20):2511–6.
Yamaguchi J, Kusano KF, Masuo O, et al. Stromal cell-derived factor-1 effects on ex vivo expanded endothelial progenitor cell recruitment for ischemic neovascularization. Circulation Mar. 2003;11(9):1322–8.
Massberg S, Konrad I, Schürzinger K, et al. Platelets secrete stromal cell-derived factor 1alpha and recruit bone marrow-derived progenitor cells to arterial thrombi in vivo. J Exp Med May. 2006;15(5):1221–33.
Engsig MT, Chen QJ, Vu TH, et al. Matrix metalloproteinase 9 and vascular endothelial growth factor are essential for osteoclast recruitment into developing long bones. J Cell Biology Nov. 2000;151(4):879–89.
Möhle R, Bautz F, Rafii S, Moore MA, Brugger W, Kanz L. The chemokine receptor CXCR-4 is expressed on CD34 + hematopoietic progenitors and leukemic cells and mediates transendothelial migration induced by stromal cell-derived factor-1. Blood Jun. 1998;15(12):4523–30.
Salcedo R, Wasserman K, Young HA, et al. Vascular endothelial growth factor and basic fibroblast growth factor induce expression of CXCR4 on human endothelial cells: in vivo neovascularization induced by stromal-derived factor-1alpha. Am J Pathol Apr. 1999;154(4):1125–35.
Saxena A, Fish JE, White MD, et al. Stromal cell-derived factor-1alpha is cardioprotective after myocardial infarction. Circulation Apr. 2008;29(17):2224–31.
Shi X, Simms KJ, Ewing TJ, et al. The bone marrow endothelial progenitor cell response to septic infection. Front Immunol. 2024;15:1368099.
Du F, Zhou J, Gong R, et al. Endothelial progenitor cells in atherosclerosis. Front Biosci (Landmark Ed) Jun. 2012;1(6):2327–49.
Hill WD, Hess DC, Martin-Studdard A, et al. SDF-1 (CXCL12) is upregulated in the ischemic penumbra following stroke: association with bone marrow cell homing to injury. J Neuropathol Exp Neurol Jan. 2004;63(1):84–96.
Stellos K, Gawaz M. Platelet interaction with progenitor cells: potential implications for regenerative medicine. Thromb Haemost Nov. 2007;98(5):922–9.
Li JH, Li Y, Huang D, Yao M. Role of stromal Cell-Derived Factor-1 in endothelial progenitor Cell-Mediated vascular repair and regeneration. Tissue Eng Regen Med. Oct 2021;18(5):747–58.
Qin G, Ii M, Silver M, et al. Functional disruption of alpha4 integrin mobilizes bone marrow-derived endothelial progenitors and augments ischemic neovascularization. J Exp Med Jan. 2006;23(1):153–63.
Hynes RO. The extracellular matrix: not just pretty fibrils. Sci Nov. 2009;27(5957):1216–9.
Karar J, Maity A. PI3K/AKT/mTOR pathway in angiogenesis. Front Mol Neurosci. 2011;4:51.
Laurenzana A, Fibbi G, Margheri F, et al. Endothelial progenitor cells in sprouting angiogenesis: proteases pave the way. Curr Mol Med. 2015;15(7):606–20.
Zhang Y, Zhang YY, Pan ZW, et al. GDF11 promotes wound healing in diabetic mice via stimulating HIF-1ɑ-VEGF/SDF-1ɑ-mediated endothelial progenitor cell mobilization and neovascularization. Acta Pharmacol Sin. May 2023;44(5):999–1013.
Li S, Tian Y, Huang X, et al. Intravenous transfusion of endothelial colony-forming cells attenuates vascular degeneration after cerebral aneurysm induction. Brain Res Dec. 2014;17:1593:65–75.
Yu G, Liu P, Shi Y, et al. Stimulation of endothelial progenitor cells by microRNA-31a-5p to induce endothelialization in an aneurysm neck after coil embolization by modulating the Axin1-mediated β-catenin/vascular endothelial growth factor pathway. J Neurosurg Sep. 2020;1(3):918–26.
Ramirez-Velandia F, Mensah E, Salih M, et al. Endothelial progenitor cells: A review of molecular mechanisms in the pathogenesis and endovascular treatment of intracranial aneurysms. Neuromolecular Med Jun. 2024;17(1):25.
Cornelissen A, Vogt FJ. The effects of stenting on coronary endothelium from a molecular biological view: time for improvement? J Cell Mol Med. Jan 2019;23(1):39–46.
Tsurumi Y, Murohara T, Krasinski K, et al. Reciprocal relation between VEGF and NO in the regulation of endothelial integrity. Nat Med Aug. 1997;3(8):879–86.
Koh GY. Orchestral actions of angiopoietin-1 in vascular regeneration. Trends Mol Med Jan. 2013;19(1):31–9.
Brindle NP, Saharinen P, Alitalo K. Signaling and functions of angiopoietin-1 in vascular protection. Circ Res Apr. 2006;28(8):1014–23.
Pang D, Wang L, Dong J, et al. Integrin α5β1-Ang1/Tie2 receptor cross-talk regulates brain endothelial cell responses following cerebral ischemia. Exp Mol Med Sep. 2018;5(9):1–12.
Ravindran K, Salem MM, Alturki AY, Thomas AJ, Ogilvy CS, Moore JM. Endothelialization following flow diversion for intracranial aneurysms: A systematic review. AJNR Am J Neuroradiol. Feb 2019;40(2):295–301.
Li ZF, Fang XG, Yang PF, et al. Endothelial progenitor cells contribute to Neointima formation in rabbit elastase-induced aneurysm after flow diverter treatment. CNS Neurosci Ther May. 2013;19(5):352–7.
Marosfoi M, Langan ET, Strittmatter L, et al. In situ tissue engineering: endothelial growth patterns as a function of flow diverter design. J Neurointerv Surg Oct. 2017;9(10):994–8.
Kalkman DN, Kerkmeijer LS, Woudstra P, et al. Three-year clinical outcomes after dual-therapy COMBO stent placement: insights from the REMEDEE registry. Catheter Cardiovasc Interv Sep. 2019;1(3):342–7.
Cortese J, Rasser C, Even G, et al. CD31 mimetic coating enhances flow diverting stent integration into the arterial wall promoting aneurysm healing. Stroke Jan. 2021;52(2):677–86.
Sun A, Huang X, Jiao Y, Wang X, Wen J. Construction of biological factor-coated stent and its effect on promoting endothelialization. Mater Sci Eng C Mater Biol Appl Mar. 2021;122:111943.
Matsuda Y, Chung J, Lopes DK. Analysis of Neointima development in flow diverters using optical coherence tomography imaging. J Neurointerv Surg Feb. 2018;10(2):162–7.
Trivelato FP, Wajnberg E, Rezende MTS, et al. Safety and effectiveness of the pipeline flex embolization device with shield technology for the treatment of intracranial aneurysms: midterm results from a multicenter study. Neurosurg Jul. 2020;1(1):104–11.
Manning NW, Cheung A, Phillips TJ, Wenderoth JD. Pipeline shield with single antiplatelet therapy in aneurysmal subarachnoid haemorrhage: multicentre experience. J Neurointerv Surg Jul. 2019;11(7):694–8.
Martínez-Galdámez M, Lamin SM, Lagios KG, et al. Treatment of intracranial aneurysms using the pipeline flex embolization device with shield technology: angiographic and safety outcomes at 1-year follow-up. J Neurointerv Surg Apr. 2019;11(4):396–9.
Zhao H, Shen S, Zhao L, Xu Y, Li Y, Zhuo N. 3D printing of dual-cell delivery titanium alloy scaffolds for improving osseointegration through enhancing angiogenesis and osteogenesis. BMC Musculoskelet Disord Aug. 2021;27(1):734.
Camci-Unal G, Nichol JW, Bae H, Tekin H, Bischoff J, Khademhosseini A. Hydrogel surfaces to promote attachment and spreading of endothelial progenitor cells. J Tissue Eng Regen Med. May 2013;7(5):337–47.
Nishi H, Ishii A, Ono I, et al. Biodegradable flow diverter for the treatment of intracranial aneurysms: A pilot study using a rabbit aneurysm model. J Am Heart Assoc Oct. 2019;15(20):e014074.
Jamshidi M, Rajabian M, Avery MB, et al. A novel self-expanding primarily bioabsorbable braided flow-diverting stent for aneurysms: initial safety results. J Neurointerv Surg Jul. 2020;12(7):700–5.
Li WD, Li XQ. Endothelial progenitor cells accelerate the resolution of deep vein thrombosis. Vascul Pharmacol Aug. 2016;83:10–6.
Yu Y, Ricciotti E, Scalia R, et al. Vascular COX-2 modulates blood pressure and thrombosis in mice. Sci Transl Med May. 2012;2(132):132ra54.
Asahara T, Takahashi T, Masuda H, et al. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. Embo J Jul. 1999;15(14):3964–72.
Cai X, Chen Z, Pan X, et al. Inhibition of angiogenesis, fibrosis and thrombosis by tetramethylpyrazine: mechanisms contributing to the SDF-1/CXCR4 axis. PLoS ONE. 2014;9(2):e88176.
Park KS, Kang SN, Kim DH, et al. Late endothelial progenitor cell-capture stents with CD146 antibody and nanostructure reduce in-stent restenosis and thrombosis. Acta Biomater Jul. 2020;15:111:91–101.
Bradbury C, Buckley T, Sun YZ, Rose P, Fitzmaurice D. Patients with high levels of Circulating endothelial progenitor cells (EPC) following at least three months of anticoagulation for unprovoked venous thromboembolism (VTE) are at low risk of recurrent VTE-Results from the exact randomised controlled trial. EClinicalMedicine Dec. 2019;17:100218.
Wei HJ, Wang D, Chen JL, et al. Mobilization of Circulating endothelial progenitor cells after endovascular therapy for ruptured cerebral aneurysms. Neurosci Lett Jul. 2011;8(2):114–8.
Wei H, Yang M, Yu K, et al. Atorvastatin protects against cerebral aneurysmal degenerative pathology by promoting endothelial progenitor cells (EPC) mobilization and attenuating vascular deterioration in a rat model. Med Sci Monit Feb. 2019;2:25:928–36.
Xu Y, Zhang B, Chen Y, et al. Simvastatin increases Circulating endothelial progenitor cells and inhibits the formation of intracranial aneurysms in rats with diet-induced hyperhomocysteinemia. Neurosci Lett Aug. 2021;24:760:136072.
Fang X, Zhao R, Wang K, et al. Bone marrow-derived endothelial progenitor cells are involved in aneurysm repair in rabbits. J Clin Neurosci Sep. 2012;19(9):1283–6.
Yu G, Liu P, Shi Y, Li S, Liu Y, Zhu W. Sitagliptin stimulates endothelial progenitor cells to induce endothelialization in aneurysm necks through the SDF-1/CXCR4/NRF2 signaling pathway. Front Endocrinol (Lausanne). 2019;10:823.
Chalouhi N, Hoh BL, Hasan D. Review of cerebral aneurysm formation, growth, and rupture. Stroke Dec. 2013;44(12):3613–22.
Yuan F, Peng W, Yang Y, et al. Endothelial progenitor cell-derived exosomes promote anti-inflammatory macrophages via SOCS3/JAK2/STAT3 axis and improve the outcome of spinal cord injury. J Neuroinflammation Jun. 2023;30(1):156.
Ma X, Wang J, Li J, et al. Loading MiR-210 in endothelial progenitor cells derived exosomes boosts their beneficial effects on Hypoxia/Reoxygeneation-Injured human endothelial cells via protecting mitochondrial function. Cell Physiol Biochem. 2018;46(2):664–75.
Li S, Wang D, Tian Y, et al. Aspirin inhibits degenerative changes of aneurysmal wall in a rat model. Neurochem Res Jul. 2015;40(7):1537–45.
Hudson JS, Marincovich AJ, Roa JA, Zanaty M, Samaniego EA, Hasan DM. Aspirin and intracranial aneurysms. Stroke Sep. 2019;50(9):2591–6.
Suzuki T, Kamio Y, Makino H, et al. Prevention effect of antiplatelets on aneurysm rupture in a mouse intracranial aneurysm model. Cerebrovasc Dis. 2018;45(3–4):180–6.
Chyatte D, Bruno G, Desai S, Todor DR. Inflammation and intracranial aneurysms. Neurosurg Nov. 1999;45(5):1137–46. discussion 1146-7.
Dastouri M, Ozdag H, Akar AR, Can A. Differentiation and molecular characterization of endothelial progenitor and vascular smooth muscle cells from induced pluripotent stem cells. Bioimpacts. 2023;13(4):289–300.
Viswanathan S, Shi Y, Galipeau J, et al. Mesenchymal stem versus stromal cells: international society for cell & gene therapy (ISCT®) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy Oct. 2019;21(10):1019–24.
ISSCR Guidelines for the Clinical Translation of Stem Cells. Curr Protoc Stem Cell Biol. Apr 2009;Appendix 1:Appendix 1B.
Acknowledgements
Not applicable.
Funding
This study was supported by the Shanghai Sailing Program (23Y1404500 to YS), National Natural Science Foundation of China (No. 82171311 and 82330039 to WZ, No. 82271339 to PL, No.82301489 to YS, No.82201462 to YL), Shanghai Excellent Academic Leader Program (No. 21XD1400600 to WZ), Special Clinical Research Project in Health Industry of Shanghai Municipal Health Commission (No. 20224Y0072 to PL), Clinical Research Plan of SHDC (No. SHDC2020CR2034B to WZ), Shanghai Municipal Health Commission (2022ZZ01006), Shanghai Municipal Health Commission(GWVI-11.1-17 to WZ).
Author information
Authors and Affiliations
Contributions
This review was completed by Shiyu Shen and Tonglin Pan. Yuan Shi conducted the literature search and wrote the literature review and manuscript. Tonglin Pan created Figs. 1 and 2. Peixi Liu, Yanlong Tian, Yuan Shi and Wei Zhu critically appraised and edited the manuscript. All authors approved the final version for submission.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
No applicable.
Consent for publication
Not applicable.
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-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.
About this article
Cite this article
Shen, S., Pan, T., Liu, P. et al. The mechanisms and applications of endothelial progenitor cell therapy in the treatment of intracranial aneurysm. J Transl Med 23, 377 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06401-w
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06401-w