Skip to main content

Unlocking the therapeutic potential of tumor-derived EVs in ischemia-reperfusion: a breakthrough perspective from glioma and stroke

Abstract

Clinical studies have revealed a bidirectional relationship between glioma and ischemic stroke, with evidence of spatial overlap between the two conditions. This connection arises from significant similarities in their pathological processes, including the regulation of cellular metabolism, inflammation, coagulation, hypoxia, angiogenesis, and neural repair, all of which involve common biological factors. A significant shared feature of both diseases is the crucial role of extracellular vesicles (EVs) in mediating intercellular communication. Extracellular vesicles, with their characteristic bilayer structure, encapsulate proteins, lipids, and nucleic acids, shielding them from enzymatic degradation by ribonucleases, deoxyribonucleases, and proteases. This structural protection facilitates long-distance intercellular communication in multicellular organisms. In gliomas, EVs are pivotal in intracranial signaling and shaping the tumor microenvironment. Importantly, the cargos carried by glioma-derived EVs closely align with the biological factors involved in ischemic stroke, underscoring the substantial impact of glioma on stroke pathology, particularly through the crucial roles of EVs as key mediators in this interaction. This review explores the pathological interplay between glioma and ischemic stroke, addressing clinical manifestations and pathophysiological processes across the stages of hypoxia, stroke onset, progression, and recovery, with a particular focus on the crucial role of EVs and their cargos in these interactions.

Introduction

Gliomas are primary brain tumors thought to originate from neuroglial stem or progenitor cells. They constitute nearly 30% of all primary brain tumors and 80–85% of malignant cases, accounting for the majority of deaths caused by primary brain tumors [1, 2]. Stroke is the second leading cause of disability and death worldwide, with the highest burden observed in low- and middle-income countries, where ischemic stroke accounts for approximately 85% of all cases [3, 4]. In this context, clinical studies have demonstrated a bidirectional relationship between glioma and ischemic stroke. The induction of ischemic stroke in glioma patients has been widely reported in clinical studies [5, 6]. On one hand, this is due to the direct infiltration or compression of blood vessels by the tumor [7]; on the other hand, the effects of paraneoplastic syndrome and anticancer treatments have also been mentioned [8,9,10]. Conversely, ischemic stroke survivors exhibit a higher age-adjusted annual cancer incidence rate compared to the general population, with evidence of spatial consistency observed [11, 12]. This bidirectional interaction between glioma and ischemic stroke, as reported in clinical studies, can be attributed to the significant overlap in their underlying pathologies, which will be illustrated in detail below.

The pathogenesis of intracranial tumors is complex, involving angiogenesis, immune suppression, metabolic alterations, genetic and epigenetic mutations, as well as changes in the tumor microenvironment and blood-brain barrier disruption [2, 13]. Ischemic stroke occurs when blood flow to a part of the brain is blocked, typically due to a blood clot or occlusion of the brain artery, which leads to a lack of oxygen and nutrients, resulting in brain cell damage [14, 15]. As the disease progresses, it involves vascular pathology, tertiary collateral circulation, tissue ischemia and necrosis, inflammatory cascades at onset, oxidative stress, disruption of the blood-brain barrier (BBB), inflammation modulation with scar formation and resolution, angiogenesis, and neural function compensation [14, 16]. It is obvious that gliomas share numerous pathological similarities with stroke, including immune modulation, oxidative stress, blood-brain barrier disruption, and angiogenesis. These mechanisms are also involved in stroke risk factors, stroke onset, and recovery.

The construction of the tumor microenvironment (TME) plays a critical role in glioma development, as the tumor modulates intracranial cell function through the TME, creating a supportive environment for its growth [17]. During this process, various bio factors, such as chemokines, are secreted via autocrine or paracrine mechanisms through extracellular vesicles (EVs), diffusing within the central nervous system [18,19,20]. EVs play a key role in complex, long-distance intercellular signaling in multicellular organisms by protecting their cargos from enzymatic degradation through a lipid bilayer structure that prevents enzyme penetration [21, 22]. Given the pivotal role of EVs in TME construction and intercellular communication, understanding the molecular interplay between glioma and ischemic stroke, particularly through the involvement of EVs, provides valuable insights into their shared pathophysiology and offers potential therapeutic targets for managing both conditions simultaneously [23,24,25].

In this review, we will elucidate the interplay between glioma and ischemic stroke, with a particular focus on EVs, which facilitate complex intercellular signaling in multicellular organisms and share numerous common cargos between glioma and ischemic stroke [26, 27].

Extracellular vesicles

EVs are small, membrane-bound vesicles secreted by cells into the extracellular environment. Since 2004, the term “exosome” has been widely used in the scientific literature to refer to various types of EVs. However, in September 2011, the International Society for Extracellular Vesicles (ISEV) formally adopted “extracellular vesicles” as a general term, leading to its widespread use [28]. This shift in terminology reflects a growing recognition that EVs are not merely a cellular waste disposal mechanism but also play a critical role in cell-to-cell communication [29].

The therapeutic and diagnostic potential of EVs arises from their ability to shield cargos during circulation and function as natural carriers of complex biological materials—including proteins, lipids, and nucleic acids (mRNA, miRNA, circRNA, lncRNA, and rRNA)—between cells [30,31,32]. Because ribonucleases, deoxyribonucleases, and proteases cannot penetrate the EV lipid bilayer, encapsulating sensitive cargos within vesicles protects it from enzymatic degradation in the extracellular environment [21, 22]. Thus, EV biogenesis represents a significant evolutionary advancement, facilitating complex intercellular signaling in multicellular organisms [26, 27]. EVs are secreted by all cell types and can be found in nearly all body fluids, including blood [33], saliva [34], cerebrospinal fluid (CSF) [35], breast milk [36], urine [37], and semen [38], underscoring their promising value of EVs in clinical biomarker studies. Their ability to travel through these fluids enables them to deliver functional information to distant sites within the body [39]. These findings confirm the unique role of EVs in cell-to-cell material transport, particularly over long distances, and provide insights into cellular processes in both pathological and physiological contexts [40].

Despite extensive research, the clear classification of EVs remains challenging. Traditionally, EVs are broadly divided into three main categories: (a) microvesicles, which are generated by outward budding and fission of the plasma membrane; (b) exosomes, formed within the endosomal system and released when multivesicular bodies fuse with the plasma membrane; and (c) apoptotic bodies, which are shed as blebs from cells undergoing apoptosis [41]. However, because assigning an EV to a specific biogenesis pathway is complex, ISEV recommends using operational terms for EV subtypes based on their physical characteristics. For example, “small EVs” (sEVs) refer to particles smaller than 100–200 nm, while “medium/large EVs” (m/l EVs) refer to those larger than 200 nm. Additionally, EVs can be categorized by density into low, middle, and high ranges, each with specific definitions [42].

EVs play a significant role in the pathogenesis of various diseases, including cancer and stroke [31, 43]. They contribute not only through intercellular communication but also by altering the extracellular environment at lesion sites and influencing both physiological and pathological metabolic processes [22, 43, 44].

Interplay revealed in clinical reports between glioma and ischemic stroke

Although ischemic stroke and glioma are separate and distinct conditions, each with its unique characteristics, numerous clinical investigations have demonstrated a distinct bidirectional relationship between glioma and stroke, as shown in Fig. 1. On the one hand, glioma can increase the risk of ischemic stroke. Certain intracranial gliomas, particularly those located in regions such as the insula, operculum, and temporal lobe, may infiltrate or compress blood vessels, potentially leading to the occurrence of ischemic stroke [7]. In a case report, two adult patients with supratentorial glioblastomas (GBM) experienced ischemic stroke precisely at the tumor site [5, 6]. A clinical study further demonstrated that patients afflicted with gliomas face an increased risk of ischemic stroke, with incidence rates up to 9%, compared to 2.7% in the general population [45, 46]. Stroke has also been identified as a common postoperative and late complication of radiotherapy, and it is linked to tumor-induced hypercoagulability or nonbacterial thrombotic endocarditis [9, 10]. In their research, Schlehofer and colleagues found a combined odds ratio of 1.9, indicating a potential association between self-reported stroke within two years before meningioma or glioma diagnosis, indicating stroke as a possible risk factor for glioma [47]. Additionally, the migration of metastatic glioma cells increases the likelihood of emboli formation, potentially leading to the onset of stroke, as revealed previously [8]. On the other hand, evidence also suggests that patients with prior stroke may have a higher risk of glioma. Wojtasiewicz et al. [12] reported a case in which a patient developed glioblastoma in a previously infarcted area, two years after experiencing an ischemic stroke. Qureshi et al. [11] recorded 3680 noncancerous adults and found that ischemic stroke survivors had a higher age-adjusted annual cancer incidence rate than the general population. A correlation has been observed between stroke and malignant glioma development. Chen et al. [48] uncovered that female patients aged 40–60 years who experienced stroke demonstrated a heightened vulnerability to glioma development, with adjusted hazard ratios of 7.41 and 16.3, respectively.

Fig. 1
figure 1

Neurovascular unit and the interplay between stroke and glioma. Numerous clinical investigations have demonstrated a distinct bidirectional enhancement between glioma and stroke, suggesting their reciprocal interaction in mechanisms. Abbreviations: ROS = reactive oxygen species, Factor X = coagulation factor X, IL-6 = interleukin-6, IL-1β = interleukin-1 beta, TNF-α = tumor necrosis factor alpha, PAI-1 = plasminogen activator inhibitor-1, EGFR = epidermal growth factor receptor, NF-kB = nuclear factor kappa-light-chain-enhancer of activated B cells, AKT = protein kinase B, MMPs = matrix metalloproteinases, SDF1 = stromal cell-derived factor 1, HIF1-α = hypoxia-inducible factor 1 alpha, VEGF = vascular endothelial growth factor

Alterations in the pathophysiological dimensions of glioma and ischemic stroke

The rapid proliferation of glioma cells, metastatic activity, blood-brain barrier disruption, and the release of micro- and macroparticles into circulation collectively contribute to thrombosis and capillary obstruction in glioma [49, 50]. This complex glioma-induced complex homeostatic changes often result in localized ischemia. Glioma cells release various procoagulant factors and cytokines, such as factor X and mucins, which activate monocytes, endothelial cells, and platelets [51, 52]. Additionally, they stimulate neutrophils to form neutrophil extracellular traps (NETs) and inhibit protein C activation [53]. These processes induce localized inflammation and ischemic hypoxia within the glioma microenvironment. Numerous studies have indicated that various forms of glioma therapy, namely platinum-based drugs, angiogenesis inhibitors, and radiotherapy, are associated with an elevated susceptibility to thromboembolism [7]. As shown in Fig. 2, we demonstrate the significant interplay between the pathophysiological dimensions of glioma and ischemic stroke, which will be described in detail in the following sections. This indicates that investigating the shared pathological mechanisms from glioma to stroke holds substantial importance in enhancing our comprehension of stroke occurrences related to cancer.

Fig. 2
figure 2

The pathological overlap between stroke and glioma is significant. Pathological interactions between glioma and stroke are highly overlapping, forming a complex network. This suggests that the impact of glioma on stroke encompasses various stages, including risk factors, pre-stroke hypoxic conditions, stroke onset, and post-stroke recovery

Hypoxia-related reactive oxygen species (ROS)

Glioma and ischemic stroke share numerous pathological pathways, including hypoxia [43]. In tumors, hypoxia can arise from either disrupted vascular supply or when tumor growth outpaces existing vascular blood support. During ischemic stroke, sudden blockage of cerebral blood flow results in rapid oxygen depletion, leading to immediate and irreversible neuronal death, accompanied by extensive brain damage at the infarct core. Numerous origins of ROS generation have been documented in both ischemia and glioma. Notably, these two conditions exhibit a shared interconnected signaling network involved in ROS production and subsequent downstream effects [7]. ROS are reactive molecules formed due to unpaired electrons in the outer orbits of specific molecules resulting from oxygen’s partial reduction [54]. This unstable state of oxygen leads to the generation of free radicals through a process of partial reduction [55].

During the progression of glioma, elevated intrinsic ROS levels are implicated in a wide array of activities, including the stimulation of oncogenes, augmentation of metabolic processes, and disruption of mitochondrial functionality [55]. In ischemic stroke, ROS levels transition from baseline to peak concentrations during the reperfusion phase [56]. This surge in ROS level potentially plays a role in processes such as apoptosis and cellular necrosis. ROS at a toxic level due to an imbalance in production and antioxidant neutralization by antioxidant enzymes, leads to cellular injury via lipid peroxidation, protein oxidation, and DNA damage [56]. Intracellular reactive ROS are generated predominantly by electron transfers from nicotinamide adenine dinucleotide phosphate (NADPH) oxidase to molecular oxygen [57].

Blood-brain barrier (BBB)

The BBB consists of the contiguous endothelial lining of capillaries, closely interlinked cellular junctions, an undamaged basement membrane, pericytes enwrapping vessels, microglial cells, and glial membrane enveloped by astrocytes [58]. Glioma advancement is closely intertwined with the development of new blood vessels, with a key clinical complication being vasogenic brain edema, which significantly elevates intracranial pressure due to compromised BBB integrity and increased permeability [59]. Glioma cells exert influences on the BBB both directly, through interactions with nearby BBB regions, and indirectly, by releasing various biochemical substances [60]. These cells secrete factors such as vascular endothelial growth factor (VEGF), which compromise BBB integrity, increase permeability, and enable tumors to access nutrients and oxygen from the bloodstream [61]. The BBB also allows immune cells, including tumor-associated macrophages, to infiltrate the brain, potentially accelerating tumor progression [62]. In the context of an ischemic stroke, disrupted BBB leads to the leakage of blood-borne cells, chemicals, and fluid into the brain parenchyma [63, 64]. This occurs due to heightened paracellular and transcellular permeability, along with significant damage to the endothelial cells that form the barrier [63, 64]. This disruption destabilizes water and ion balance in the brain, resulting in cerebral edema [64, 65]. The infiltration of leukocytes further exacerbates inflammation, intensifying damage to brain tissue [64, 66]. In stroke or glioma, although BBB dysfunction generally leads to adverse outcomes, it may offer a potential advantage by allowing therapeutic agents to reach intended brain targets more effectively.

Neurovascular unit

Pericytes

Pericytes envelop a significant portion of the surface of brain capillary endothelial cells, especially in regions with a pericyte-to-endothelial cell ratio of 1:3 [67]. As essential components of the neurovascular unit and BBB, pericytes act as gatekeepers, regulating the passage of brain cells as well as the transport of nutrients and other substances between the bloodstream and the brain’s interstitial fluid [68]. Numerous studies suggest an association between pericytes and pathological changes in the BBB, which have been linked to stroke. Ischemic injury to pericytes within the cerebral microvasculature has a detrimental effect on the damage caused by stroke and the development of brain edema [69, 70]. This injury disrupts microvascular blood flow and compromises the integrity of the blood-brain barrier, exacerbating the overall impact of the stroke on brain tissue [69, 70]. Conversely, signaling pathways activated in pericytes within the vasculature of the peri-infarct area in response to ischemia positively impact stroke outcomes. These signaling events promote post-stroke angiogenesis and neurogenesis, contributing to recovery and healing after a stroke [70, 71]. While in the glioma context, glioblastoma cells leverage interactions with pericytes to promote GBM cell proliferation and enhance tumor growth [72]. An in vivo investigation of glioblastoma cell proliferation was conducted by grafting co-cultured human RFP-labeled glioblastoma cells and GFP-labeled mouse pericytes onto the brain cortex of an immunocompetent mouse model. Mice with these xenografts exhibited an enhanced level of perivascular infiltration of glioblastoma cells in their brain tissues [73]. Pericytes play crucial roles in tumor angiogenesis. Huizer et al. indicated that pericytes serve as the primary source of periostin in human gliomas, where periostin plays a vital role in facilitating blood vessel growth and branching [74]. Furthermore, pericytes can modulate their own immune properties, enabling immune evasion. In vitro, analysis of membrane molecules involved in suppressing antitumor immune responses, such as interleukin-1 receptor antagonists, revealed that pericytes display an immunosuppressive pattern of surface molecules after interacting with glioblastoma cells [72].

Astrocytes

Astrocytes, especially their extended endfeet encompassing the extracellular matrix and pericytes, serve as the final barrier against the entry of undesirable proteins and molecules. These astrocyte-loaded endfeet al.so play a crucial role in strengthening the BBB by releasing specific bioactive substances including growth factors such as VEGF and glial cell-derived neurotrophic factor [75,76,77]. Astrocytic dysfunction, marked by the detachment of endfeet from the basement membrane, occurs shortly after a stroke [77, 78]. In response to minor ischemic injury, astrocytes begin proliferating, causing enlargement of both their cell bodies and processes along with elevated expression of glial fibrillary acidic protein (GFAP). As the extent of injury escalates, astrocytes undergo substantial proliferation, resulting in a more pronounced increase in GFAP expression. Glioma cells establish attachments to blood vessels utilizing bradykinin, a chemotactic signaling peptide produced by vascular endothelial cells [79]. Glioma cells then proceed to envelop the external surface of the existing blood vessels through invasive and parasitic mechanisms, infiltrating the adjacent space [80]. This invasive conduct disrupts the connection between astrocyte end feet and endothelial cells, which ultimately results in the breakdown of the BBB [81]. Notably, ischemia-induced astrocyte activation, followed by mutations in genes like neurofibromatosis type 1 and glycoprotein podoplanin during reactive gliosis, has been implicated in gliomagenesis [7]. This is due to the proposition that both glial progenitor cells and reactive astrocytes are suggested sources of identical lineages. Reactive astrocytes also enhance glioma cell proliferation and migration by producing matrix metalloproteinases (MMPs) and releasing stromal cell-derived factor 1 (SDF1) [82, 83]. Furthermore, tunneling nanotubes and the secretion of various molecules such as Interleukin-6 (IL-6), IL-19, Insulin-like Growth Factor 1 (IGF-1), transforming Growth Factor-β (TGF-β), monocyte Chemoattractant Protein 4 (MCP4), and VEGF also contribute to the role of reactive astrocytes in promoting glioma infiltration [84, 85].

Microglia

In a normal physiological state, microglia, the resident immune cells of the central nervous system (CNS), exhibit a ramified morphology and contribute significantly to brain homeostasis, thus forming an integral component of the neurovascular unit [86]. Upon exposure to environmental stress or injury, microglia transform morphologically, adopting an amoeboid shape with shorter projections and larger cell bodies. The diverse biological characteristics exhibited by microglia in response to injury correspond to distinct phenotypes, as indicated by the pro-inflammatory M1 phenotype and the anti-inflammatory M2 phenotype [87]. These phenotypes are marked by unique secretion patterns. M1 microglia release pro-inflammatory cytokines, including IL-6, IL-1β, ROS, tumor necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS), and reactive nitrogen species (RNS). In contrast, M2 microglia predominantly secrete anti-inflammatory cytokines such as IL-4, IL-10, TGF-β1, IGF-1, and nerve growth factor (NGF) [88, 89]. Manipulating the transition of resident microglia from the M1 to the M2 state presents an intriguing target for potential therapeutic interventions in stroke treatment [90].

In glioma, microglia and macrophages constitute the most abundant population of infiltrating cells, collectively accounting for at least one-third of the total tumor mass [91]. Evidence supports that microglia significantly contribute to creating a tumor-promoting microenvironment that promotes the growth of gliomas [92]. Specifically, the presence of transformed microglia, transitioning from the M1 (tumor-suppressive) to the M2 (tumor-promoting) state, is a key facilitator in inducing immune suppression within the tumor region [7]. This transformation and immune suppression contribute to the augmentation of tumor expansion, metastasis, angiogenesis, and the maintenance of glioma stem cells. These processes are mediated through the secretion of various factors, such as MMPs, VEGF, IL-10, TGF, TNF, chemokine ligand 18 (CCL18), and CCL22, CXCL12, and Fas ligand [7].

Angiogenesis

Recent studies have highlighted the importance of neurovascular networks, underscoring the critical communication between neurons and blood vessels for proper brain function [93]. Sprouting angiogenesis, which involves the emergence of new blood vessels from existing ones, plays a crucial role in both physiological processes such as tissue regeneration, wound healing, and morphogenesis, as well as in pathological conditions like tumor growth and ischemic stroke [94]. Although both stroke and tumor angiogenesis involve the formation of new blood vessels, their underlying mechanisms, purposes, and outcomes are distinct. Poststroke angiogenesis is generally regarded as a favorable defense response against hypoxia by improving the blood supply to the brain tissue. In contrast, increased angiogenesis within cancer tissue supplies oxygen and nutrients to cancer cells, thereby promoting tumor growth, invasion, and metastasis [95].

Angiogenesis typically begins 4 to 7 days after cerebral ischemia, primarily at the boundary between the ischemic core and the surrounding tissue. This post-ischemic angiogenesis potentially supports the remodeling of neurons by promoting neurogenesis and participating in the guidance of sprouting axons, facilitated by signaling pathways involving vascular endothelial growth factor and laminin/β1-integrin [96]. In tumors, the expanding cell mass leads to both an increased oxygen demand and an increased diffusion distance between nearby capillaries and the central region, which together contribute to decreased oxygen availability [97]. This phenomenon further contributes to the establishment of a hypoxic microenvironment. Cancer angiogenesis is then activated to meet the nutritional and oxygen demands of cancer cells, enabling their proliferation. This process includes multiple stages: basement membrane degradation, endothelial cell proliferation, migration, sprouting, branching, and tubular structure formation [43, 98].

Cancer cells produce a variety of proangiogenic factors while concurrently suppressing the action of antiangiogenic factors within their surrounding environment, thereby facilitating the growth of blood vessels [43]. It’s important to mention that certain tumor cell secretions, such as extracellular vesicle-associated miR-181b-5p and miR-210, have been reported to promote post-stroke angiogenesis [43]. These findings suggest the therapeutic potential of glioma-derived EVs through post-stroke angiogenesis. However, comprehensive information regarding these phenomena is currently limited, and further substantiated evidence is required to fully understand and validate these aspects.

Immune response

The immune response is divided into two main branches: the innate and adaptive immune systems. The innate immune response, also known as the inflammatory response, acts as the body’s first line of defense against pathogens and is the primary reaction to tissue injury [99]. In contrast, the adaptive immune system, involving lymphocytes (B cells and T cells), is more specialized and targets specific pathogens. While the adaptive response takes longer to activate initially, it develops immunological memory, allowing for a faster and more efficient reaction upon re-exposure to the same pathogen [99]. Inflammation can exacerbate tissue damage in acute stroke, but subsequent adaptive immune responses and anti-inflammatory processes are essential for tissue repair [14, 100]. After a stroke, damaged cells release alarm signals or molecules referred to as damage-associated molecular patterns (DAMPs), which encompass a variety of components, such as nuclear proteins, nucleic acids, heat-shock proteins, and other molecules [101]. DAMPs act as initiators, activating immune responses by binding to specialized pattern recognition receptors. Glioma cells, on the other hand, have the ability to attract various cells, including immune cells, to their microenvironment by releasing cytokines such as TGF-β and GM-CSF which serve as signals that guide these immune cells to the specific niche [102, 103]. Subsequently, the glioma cells influence these recruited cells, inducing them to adopt tumor-promoting phenotypes that facilitate the tumor’s growth and progression [104]. Once recruited, these immune cells can form a physical barrier, impeding the access of additional immune cells and hindering their ability to target tumor cells effectively. Glioma-associated microglia and macrophages can drive immune cells into pro-inflammatory or anti-inflammatory phenotypes, modulate the immune response, and reduce the immune system’s attack on tumor cells, thereby enhancing glioma survival [105]. Additionally, glioma-associated microglia and macrophages reorganize the extracellular matrix, making tumor cells more susceptible to invasion [106, 107]. In conclusion, in ischemic stroke, the immune response plays a dual role: it aims to repair damaged tissue and restore homeostasis, while also potentially exacerbating injury through inflammatory processes [100]. In contrast, the immune response in tumors focuses primarily on recognizing and eliminating abnormal cells within the complex tumor microenvironment [104, 105].

The impacts of diverse cargos within glioma-derived EVs on glioma progression

The tumor microenvironment comprises tumor cells and surrounding components, including innate and adaptive immune cells, mesenchymal fibroblasts, and vascular and lymphatic networks [17]. Various chemokines, secreted through autocrine or paracrine mechanisms, make up the TME [18, 19]. Tumor growth is driven by the interactions between tumor-residing cells and the TME, and alterations in the microenvironment can significantly affect tumorigenesis and progression [17, 25]. Glioma-derived EVs transport bioactive cargos into the TME and recipient cells, playing critical roles in intercellular communication [24, 108]. These EVs can alter cellular functions or reprogram recipient cells, influencing tumor immune tolerance, promoting malignant transformation, and mediating interactions within the TME. Additionally, glioma-derived EVs regulate glioma cell stemness, contribute to angiogenesis, drive treatment resistance, and are implicated in neurodegenerative disease pathology [24, 108].

The vesicles carry a range of molecular modifiers that facilitate communication between cancer cells and surrounding stromal cells, with numerous EV cargos identified in clinical reports [109,110,111]. This EV-mediated intercellular communication provides critical insights into the molecular characteristics of tumors, promoting tumor growth, metabolism, invasion, and resistance [23, 112]. Beyond their direct impact on glioma properties, EV cargos also have indirect effects, including immune suppression and angiogenesis. In this review, we compile recent reports on glioma-derived EV cargos, as shown in Table 1. We categorized them by function and ranked them based on chemical properties, such as proteins and nucleic acids.

Table 1 Cargos within glioma-derived EVs

Glioma-derived EVs participate in constructing an immunosuppressive TME

In the immunosuppressive tumor microenvironment, immune cells and glioma cells compete for nutrients, and the metabolites produced during this competition can influence immune cell differentiation and function [17, 113]. This competition contributes to the immunosuppressive environment, where tumor-released cargos in EVs transmit signals that are taken up by immune cells in the brain, shifting their cytokine profiles toward immune suppression [114, 115]. In this process, the recipient cells of these cargos include monocytes, myeloid-derived suppressor cells (MDSCs), NK cells, CD8 + T cells, CD4 + T cells, microglia, and macrophages, often resulting in the suppression of immune cell activity, induction of M2 macrophage polarization, regulation of microglia, activation of MDSCs, and modulation of T cell expansion or function. Ultimately, these interactions facilitate metastasis and immune evasion [116,117,118,119,120,121,122,123]. Though immunosuppressive cargos are predominant, immune-activating cargos have also been identified. For instance, Shah et al. found that miR-1983, a glioma-derived cargo, enables NK cells to kill glioma cells through the miR-1983-TLR7-IFNβ circuit [120].

Glioma-derived EVs enhance glioma-related angiogenesis

Tumor-derived EVs have been reported to facilitate angiogenesis by delivering functional and regulatory factors that induce proangiogenic or anti-apoptosis changes in brain microvascular endothelial cells [124, 125]. GBM-derived EVs can not only transfer mRNAs into endothelial cells, leading to transcriptional changes and the subsequent synthesis of functional proteins in these recipient cells [126]. These vesicles are also enriched in angiogenic protein factors and promote tubule formation [127, 128]. Interestingly, the proteome and mRNA profiles of EVs closely reflect the oxygenation status of donor glioma cells [124]. EVs are also enriched with angiogenesis-related regulatory factors and are released with increased autocrine and promigratory activation of GBM cells [129]. Recent research has identified long non-coding RNAs, such as CCAT2 and POU3F3, as enhancers of angiogenesis [125, 130]. Conversely, EV-derived miR-1 and Annexin A2 (ANXA2) have been reported as orchestrators of neovascularization, targeting multiple pro-oncogenic signals [131].

Glioma-derived EVs represent a promising field in clinical biomarker studies

Due to their intracranial location, glioma tissues are less accessible than other solid tumors, making the identification of prominent biomarkers in biological fluids essential. Given the unique characteristics of the blood-brain barrier, studies of cerebrospinal fluid hold particular significance for gliomas compared to other tumors [126]. Following this perspective, numerous studies on CSF and serum have been conducted, highlighting glioma-derived EVs due to their enrichment of specific factors [35]. Particularly following the extensive application of proteome profiling, genomics, and microarray analysis, various biomarkers, such as epidermal growth factor receptor variant III (EGFRvIII) and miR-21, have been widely studied within glioma-derived EVs and found in CSF and plasma [132,133,134].

Potential effects of cargos in glioma-derived EVs on ischemic stroke

The pathogenesis of ischemic stroke is complex and multifactorial, with common etiologies including large artery atherosclerosis, cardioembolism, and small artery occlusion [135, 136]. Progression of ischemic stroke involves vascular pathology, formation of tertiary collateral circulation, tissue ischemia and necrosis, inflammatory cascades, oxidative stress, blood-brain barrier disruption, scar formation and resolution, and neural function compensation [14, 16]. Hemorrhagic stroke, on the other hand, primarily involves mass effect and hematoma absorption, both significantly influenced by inflammation [137]. As shown in Fig. 3, glioma-derived extracellular vesicles carry cargos originating from the tumor microenvironment and spread intracranially. Notably, these cargos have been widely implicated in stroke, where they exhibit coherent roles in both conditions, as detailed in the following sections. Given the efficiency and ubiquity of EVs in intercellular communication and their role in pathological processes like inflammation and angiogenesis, we hypothesize that glioma-derived EVs may provide a critical link between prevalent neurovascular diseases and intracranial tumors.

Fig. 3
figure 3

Glioma-derived EVs and their role in stroke. Glioma-derived extracellular vesicles carry cargos that have been widely reported in stroke and play coherent roles in both conditions. Abbreviations: IL-6 = interleukin-6, SDF-1α = stromal cell-derived factor 1 alpha, IFN-γ = interferon gamma, PD-L1 = programmed cell death ligand-1, L1CAM = neural cell adhesion molecule L1, TrkB = tropomyosin receptor kinase B, AQP4 = aquaporin 4, HIF-1α = hypoxia-inducible factor 1 alpha, VEGF = vascular endothelial growth factor, PDGF = platelet-derived growth factor, MMPs = matrix metalloproteinases, CXCR4 = C-X-C chemokine receptor type 4

Glioma-derived EV cargos associated with tumor growth, metabolism, invasion, and metastasis, and their effects on ischemic stroke

An important aspect of glioma growth and invasion is the communication and manipulation of other cells within the brain microenvironment, which supports tumor progression and resistance to therapy. In this process, extracellular vesicles play a crucial role [25, 138]. In addition to their indirect effects on immune suppression and angiogenesis, glioma-derived EVs directly impact glioma characteristics by promoting motility, proliferation, and invasiveness [138]. Notably, as shown in Table 2, some EV cargos commonly observed in ischemic stroke pathology also play similar roles in glioma. For example, Pace et al. demonstrated through in vitro experiments that L1 cell adhesion molecule-decorated exosomes increase the motility, proliferation, and invasiveness of cell lines [112]. Interestingly, L1CAM has also been identified as a biomarker in stroke patients [139]. Similarly, Colombo et al., using wild-type and tropomyosin receptor kinase B-deficient astrocytes, found that astrocyte-derived tropomyosin receptor kinase B (TrkB) expression is upregulated in stroke, promoting Aquaporin 4 (AQP4) upregulation via hypoxia-inducible factor 1 alpha (HIF1-α) activation under hypoxic conditions. This contributes to brain injury and edema formation [140]. Notably, these same factors—TrkB, AQP4, and HIF1-α—are also identified as EV cargos in glioma, where they significantly contribute to tumor cell proliferation, migration, and resistance [141,142,143]. This section will provide a detailed discussion of these individual cargos.

Table 2 Cargos associated with tumor growth, metabolism, invasion, and metastasis, along with their roles in stroke

The enhancing or suppressing roles in cellular activity and behavior are generally consistent in both stroke and glioma contexts. For example, TrkB exhibits an enhancing role in both contexts. Studies using YKL-40-silenced glioblastoma cells have highlighted that TrkB-containing exosomes contribute to glioblastoma by promoting tumor cell proliferation and activating endothelial cells [141]. In stroke, TrkB’s role in neurological recovery is well-documented, where it enhances corticospinal synaptic connections and neuroplasticity, thus improving patient outcomes [144,145,146]. In contrast, MiR-375 functions as a suppressor in cellular activities within both glioma and stroke. In glioma, MiR-375, a cargo of glioma-derived EVs, is found to play a protective role by suppressing glioma proliferation, migration, and invasion [147]. In stroke, downregulation was found in patients with ischemic stroke and in rat models, and protection of MiR-375 was approved in stroke which may achieved by reducing apoptosis and oxidative stress [148]. AQP4, another cargo found in both glioma and stroke, is released through EVs in glioma and influences the glioblastoma microenvironment by promoting a migratory phenotype in adjacent tumor cells [142]. While AQP4 plays a complex, dual role in stroke, where it exacerbates cerebral edema formation in the early stages but mitigates it in later stages [149, 150]. Lastly, miR-148a, a glioblastoma proliferation-promoting cargo, has also been identified as a biomarker for ischemic stroke in blood samples [151,152,153].

In gliomas, the influence of EV cargos on proliferation and invasiveness frequently coincides with the development of treatment resistance. For example, in the radioresistant U251 glioma cell line, circATP8B4 has been identified as a radioresistance-related cargo and is associated with enhanced glioma cell proliferation, motility, and invasion [154, 155]. Temozolomide resistance in gliomas is also usually accompanied by tumor proliferation, growth, and invasion, which has been extensively studied due to its clinical significance. Numerous temozolomide resistance-related cargos have been identified, including miR-221, miR-151a, miR-30b-3p, and lncRNA SBF2-AS1 [156,157,158]. While miR-221 and miR-151a are also recognized in the pathology of stroke [159, 160]. Using the U87-MG glioblastoma cell line, glioblastoma-derived EVs carrying HIF-1α were shown to promote tumor progression and radioresistance [143]. The expression of HIF-1α induced by stroke is time-dependent, and plays an extensive role in the pathophysiology of stroke, including neuronal survival, neuroinflammation, angiogenesis, glucose metabolism, and blood-brain barrier regulation [161,162,163]. Through experiments involving lentivirus-infected mice and primary brain cells, Cui et al. demonstrated HIF-1α’s modulatory effects on brain injury and neuroinflammation [164]. The protective function may arise from both non-transcriptional mechanisms and its function as a nuclear transcription factor [165].

Glioma-derived EV cargos associated with angiogenesis and their effects on ischemic stroke

Angiogenesis plays a crucial role after stroke and indicates a better outcome [16, 166, 167]. This is because neurogenesis and angiogenesis are highly coupled, working together to create restorative microenvironments within ischemic tissue, which leads to improved neurological function [168, 169]. Interestingly, angiogenesis can develop before stroke onset as a structural and functional adaptation to hypoxia, which often occurs due to chronic conditions like atherosclerosis or Moyamoya disease [170,171,172]. Key biomarkers, such as VEGF, MMPs, HIF-1α, TGF-β1, IL-6, and IL-8, serve as indicators of new blood vessel formation in stroke [127, 128, 173]. In the meantime, they are also angiogenesis-promoting cargos widely reported in glioma-derived EVs [174]. Similarly, as shown in Table 3, a large number of angiogenesis-related cargos from glioma-derived EVs overlap with angiogenesis-promoting factors in ischemic stroke. Due to the significant overlap in the underlying mechanisms and involved biological factors, it is reasonable to hypothesize that cargos from glioma may influence angiogenesis in the context of stroke.

Table 3 Cargos associated with angiogenesis and their roles in ischemic stroke

Using wound closure assay and tube formation assay, Giusti et al. demonstrated a dose-dependent effect of glioblastoma-derived EVs on endothelial growth [174]. In the current study, many angiogenesis-related cargos were identified in these EVs, including plasminogen activators (tPA and uPA), PAI-PA, CXCR4, VEGF, MMPs (MMP-2 and − 9), and TGF-β, most of which are also crucial angiogenic factors in stroke [174]. In the ischemic stroke context, the fibrinolytic system plays an important role in angiogenesis, in which tissue plasminogen activator (tPA) and urokinase plasminogen activator (uPA) function as plasminogen activators, promoting fibrinolysis by converting plasminogen to plasmin. While plasminogen activator inhibitor-1 (PAI-1) serves as an inhibitor by forming inhibitory complexes (PAI-PA) with tPA and uPA, thereby maintaining the balance between coagulation and fibrinolysis [175, 176]. CXCR4 typically serves as the receptor in the SDF-1/CXCR4 pathway and plays an important role in neuroprotection and angiogenesis after stroke [177,178,179]. Additionally, CXCR4 is involved in the migration of stem cells to infarcted brain areas, promoting recovery after stroke [180]. The VEGF family is well known for regulating vascularization. In the brain, VEGFs are crucial for angiogenesis, neuroprotection, and neurogenesis [181, 182]. Matrix metalloproteinases, key extracellular endopeptidases, digest extracellular proteins and serve as biomarkers for stroke. Among them, gelatinases (MMP-2 and MMP-9) are the most studied and can promote neurological recovery via angiogenesis in stroke [183, 184]. Lastly, using an oxygen-glucose deprivation model, Zhang et al. found that EVs from hypoxia-preconditioned microglia promote angiogenesis through the TGF-β/Smad2/3 pathway [185].

Using both in vivo and in vitro models, Kucharzewska et al. demonstrated alterations in the proteome and mRNA profiles under hypoxic conditions. Compared to normoxic exosomes, hypoxic exosomes were found to correlate with tumor vascularization, pericyte vessel coverage, glioblastoma cell proliferation, and decreased tumor hypoxia in a mouse xenograft model [129]. Their study identified caveolin-1, PTX3, IL-8, MMP9, platelet-derived growth factors (PDGFs), CD26, PAI1, and lysyl oxidase as cargos of glioma-derived EVs [129]. In the context of stroke, these cargos are similarly involved. Blochet et al. observed increased caveolin-1 expression in new blood vessels within the lesion and in reactive astrocytes in peri-lesion areas, promoting neovascularization, astrogliosis, and scar formation [186]. Rajkovic et al. reported significantly decreased vessel diameter, vessel proliferation, vascular density, and reactive astrocytes in PTX3 knockout (KO) mice, revealing the angiogenic role of PTX3 in stroke [187]. The PDGF family, well-studied in vascular physiology and pathology, including angiogenesis, is also relevant in stroke [188]. Elevated levels of VEGF in stroke are associated with better outcomes, potentially due to the angiogenic role of PDGF [189,190,191]. Moreover, human bone marrow mesenchymal stem cells derived VEGF were found to be stimulated by IL-8 after stroke. IL-8 is an inflammatory chemokine with potent proangiogenic properties in stroke and also serves as angiogenesis-related cargo from glioma EVs [129, 192]. Additionally, IL-6, another inflammatory chemokine found in EVs from glioma, is involved in microglia-mediated vascular repair and functional recovery after cerebrovascular injury [193, 194].

Non-coding RNAs (ncRNAs), when acting as cargos of glioma-derived EVs, exhibit significant overlap in angiogenesis-related functions between gliomas and indicators of vascular development in stroke. ncRNAs, including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are functional RNA molecules that regulate the expression and function of various genes through different mechanisms [195,196,197]. Glioma-derived EVs contain microRNAs such as miR-29a, miR-30e, miR-26a, and miR-1, which have been identified as pro-angiogenic microRNAs [126, 128, 131]. In addition to microRNAs, long non-coding RNA HOTAIR has been identified as a cargo of glioma-derived EVs by Ma et al. It was found to enhance angiogenesis by induction of VEGFA expression in glioma cells and facilitating its transmission to endothelial cells via glioma cell derived-extracellular vesicles [198]. Regarding the role of ncRNA cargos in stroke, various non-coding RNAs (ncRNAs) have been identified as key players with studies demonstrating that changes in their expression levels during and after stroke can significantly influence angiogenesis [195, 199]. The microRNA cargos from glioma-derived EVs, previously mentioned, including miR-26a and miR-210, also play similar roles in promoting angiogenesis in stroke [200,201,202]. Lastly, using an oxygen-glucose deprivation/reperfusion model of human brain microvascular endothelial cells, lncRNA HOTAIR was identified as a common factor in both glioma-derived EV cargos and as a mediator of angiogenesis in hypoxic-ischemic conditions [203].

Glioma-derived EV cargos associated with inflammation and their effects on ischemic stroke

In the brain, the interaction between immune system activation and inflammation exerts both harmful and protective effects on central nervous system function [204, 205]. In stroke, although inflammatory cascades can exacerbate tissue damage in the acute phase, adaptive immune responses and inflammatory processes are essential for subsequent tissue repair [14]. Phagocyte activity exemplifies this dynamic: microglial cells, macrophages, and phagocyte-mediated inflammatory responses play pivotal roles in the onset, progression, and outcomes of brain injury following ischemic stroke [206]. In this process, the polarization of microglia or macrophages into the M1 phenotype typically results in the release of destructive pro-inflammatory mediators, whereas M2 polarization clears cellular debris through phagocytosis and releases numerous protective/trophic factors [100]. Due to this dualistic nature, subtle modulation is critical for maintaining the balance between these phenotypes in stroke [100]. The situation is different for glioma, as the formation of an immunosuppressive tumor microenvironment is essential. The cargos released by glioma cells generally contribute to the immunosuppression of immune cells, which is critical for tumor proliferation [17]. Thus, M2 macrophage polarization, rather than M1, becomes significant, promoting tumor growth, invasion, and even angiogenesis; consequently, glioma-derived EV cargos frequently drive macrophages toward an M2 phenotype [193]. Using in vivo and in vitro models, van der Vos et al. directly visualized the release of EVs from glioma cells and their uptake by brain microglia and monocytes/macrophages, which led to increased microglial proliferation and a cytokine profile shift toward immune suppression [114]. In the current study, miR-451 and miR-21 have been identified as EV cargos, where they are also recognized as protective agents in ischemic stroke, potentially by regulating apoptosis [207,208,209,210].

As shown in Table 4, the overlap of biofactors is evident in the polarization of phagocytes (microglia and macrophages), both of which participate in immune modulation in glioma and ischemic stroke. For example, in glioma, Qian et al. discovered an enrichment of miR-1246 in the cerebrospinal fluid of GBM patients, which decreased after tumor resection. Their study found that hypoxic glioma-derived exosomes (H-GDEs) significantly promote M2 macrophage polarization, with miR-1246 identified as the most enriched microRNA in H-GDEs through microRNA sequencing analysis [123]. Notably, miR-1246 has also been identified as a potential diagnostic biomarker for ischemic stroke in serum [211]. Additional polarization-related cargos in glioma-derived EVs include IL-6 and miR-155-3p, which promote M2 macrophage polarization via the IL-6-pSTAT3-miR-155-3p-autophagy-pSTAT3 positive feedback loop, further enhancing glioma progression [193]. Meanwhile, IL-6 is critical in stroke for modulating immune responses and the acute phase reaction [212]. Interestingly, in turn, these tumor-associated macrophages can also produce EVs decorated by immunosuppressive and tumor-growth-promoting proteins, which can promote tumor cell migration and proliferation [213].

Table 4 Cargos associated with inflammation and their roles in ischemic stroke

The overlap extends to myeloid-derived suppressor cell-related EV cargos in glioma and inflammation modulators in ischemic stroke, as shown in Table 4. Myeloid-derived suppressor cells (MDSCs) belong to a key cell population responsible for regulating immune responses and are highly effective at suppressing T cell function, and play a crucial role in both tumor progression and stroke [214, 215]. Using glioma cell lines, Zhang et al. found that basic leucine zipper ATF-like transcription factor 2 (BATF2) inhibits intracellular SDF-1α production, reducing SDF-1α levels in EVs and subsequently inhibiting the recruitment of MDSCs [216]. While in stroke, SDF-1α also modulates inflammation. As a chemokine, SDF-1α is crucial for monocyte homing and migration across the blood-brain barrier, playing a significant role in the late infiltration of the penumbra by recruiting monocytes to infarcted tissue in the later stages of stroke [217, 218]. Not only chemokine, miRNAs, including miR-1246, miR-1298-5p, miR-29a, miR-92a, miR-10a, and miR-21, were found to be enriched in the body fluids of glioma patients and shown to drive the activation of MDSCs through different pathways, some of which are also matters in stroke [119, 219, 220]. In stroke, elevation of miR-29a has been shown to have a protective function by controlling M1 microglia pre-polarization or by promoting axonal outgrowth and neurological recovery [221,222,223]. Protection function was also found in miR-10a by inhibiting atherosclerotic lesion formation, and in miR-92 by alleviating cerebral vascular injury [148, 224, 225]. Notably, miR-21, a glioma EV biomarker, has been detected in CSF and serum of glioma patients and correlates with poor prognosis and tumor recurrence [133, 226,227,228]. It plays an important role in maintaining the immunosuppressive environment in glioma, possibly mediated by MDSCs, and has an anti-apoptotic effect [114, 119]. MiR-21 also acts as an inflammation modulator in stroke and has been consistently shown to be a potent anti-apoptotic factor, playing a neuroprotective role in cerebral ischemic and reperfusion injury [209, 229, 230]. Additionally, miR-21 shows potential for promising clinical outcomes [208, 231, 232].

T cell-related glioma cargos similarly overlap with stroke-related inflammatory factors, as shown in Table 4. Liu et al. found, using the GL26 cell line, that glioblastoma-derived exosomes reduce CD8 + T cell numbers and functionality, fostering tumor growth in an immunosuppressive tumor microenvironment. IFN-γ and granzyme B were identified as key cargos in these EVs during this period [121]. IFN-γ serves as an indicator of inflammation in stroke, especially of T-cell function and stroke-associated infections, it can also serve as a protective biomarker in the clinic, which was reported to elevate in stroke patients [233,234,235,236,237]. Ricklefs et al. demonstrated that glioblastoma-derived EVs can suppress T cell activation and proliferation through T cell receptor interaction with anti-CD3, or alternatively, by antigen presentation via dendritic cells (DCs). In their study, PD-L1 was found on the surface of some glioblastoma-derived EVs, both PD-L1 and PD-L1 DNA were identified as cargos of glioma-derived EVs in the process of T cell inhibition [238]. In stroke, the T-cell regulatory role of PD-L1 has also been observed. Kim et al. demonstrated that soluble PD-L1 binds to PD-1, redirecting monocyte fates from pro-inflammatory, classical phenotypes to non-classical, anti-inflammatory ones, thereby protecting against damage after middle cerebral artery occlusion [239]. Beyond its immunosuppressive role, PD-L1 was also found to regulate platelet activation and thrombosis through the Caspase-3/GSDME pathway in stroke [240]. Conversely, a study by Bodhankar et al. challenges these findings, suggesting that PD-L1 may worsen stroke outcomes, possibly through effects on suppressor T cells [241]. Studies involving patient clinical samples have identified CD73 as a crucial cargo in glioblastoma EVs, capable of being taken up by T cells and inhibiting their clonal expansion in vivo [242]. CD73 has a similar function in stroke, where it regulates leukocyte (including T cell) trafficking in the ischemic brain and serves as an outcome indicator in MCAO mice [243, 244].

Retrospect and prospect

We have discussed the pathological overlap between glioma and ischemic stroke. The interplay between these two conditions is complex, particularly with the involvement of extracellular vesicles, which efficiently facilitate intracranial interactions. However, the precise influence of glioma and glioma-derived EVs on the entire ischemic stroke process remains unclear. We aim to speculate on the impact of glioma as a risk factor for stroke, as well as its effects during the pre-stroke phase, stroke onset, and post-stroke recovery.

Glioma can be a risk factor for stroke. As an intracranial tumor, its direct infiltration or compression of blood vessels is evident and commonly observed in clinical practice [7]. Additionally, the disruption of blood vessels may lead to hemorrhagic stroke in glioma patients [245]. Chronic activation of the coagulation system, particularly as part of paraneoplastic syndrome, represents another mechanism by which glioma acts as a risk factor for ischemic stroke [246]. Coagulation abnormalities are also a frequent postoperative and late complication of radiotherapy, contributing to stroke risk through tumor-induced hypercoagulability or nonbacterial thrombotic endocarditis [9, 10].

Before the onset of stroke, chronic hypoxia and angiogenesis may have already occurred. It is well established that tumor hypermetabolism increases the demand for nutrients and oxygen [97, 170,171,172]. Collateral circulation and angiogenesis are likely enhanced in the tumor’s hypoxic microenvironment, facilitated by angiogenesis-related cargos from EVs. In the case of ischemic stroke onset, as discussed previously, the inflammatory cascade contributes to tissue damage by enlarging the infarct area, increasing blood-brain barrier permeability, and worsening cerebral edema. However, the immunosuppressive microenvironment created by glioma is speculated to provide a protective function during ischemic stroke [247]. This protective effect may extend into the recovery phase, as M2 polarization—an essential feature of the glioma-associated immunosuppressive microenvironment—helps clear cellular debris via phagocytosis and releases protective/trophic factors that promote angiogenesis and neurogenesis [100, 248]. Additionally, glioma-derived EVs carry cargos that overlap with angiogenesis promoters are presumed to involve in stroke recovery.

In the context of clinic therapy, the simultaneous occurrence of glioma and ischemic stroke presents significant challenges in stroke management. On one hand, glioma’s direct invasion exacerbates brain tissue damage, compresses and invades blood vessels, competes for oxygen and nutrients, and worsens cerebral edema. On the other hand, glioma-induced coagulation abnormalities and vascular disruption complicate clinical anticoagulation or antiplatelet therapies, increasing the risk of intracranial hemorrhage.

Understanding the molecular interplay between glioma and stroke, particularly through the involvement of EVs, offers new insights into their shared pathophysiology and provides potential therapeutic targets for managing both conditions simultaneously. However, further research is needed. The impact of glioma-derived EVs on ischemic stroke progression has not been fully confirmed, and the use of dual rodent models of glioma and stroke could be instrumental in investigating this interaction. Lipidomic analysis or specific labeling could help determine whether glioma-derived EVs play a role in the stroke-affected region. As discussed, stroke presents distinct characteristics in different phases, and the impact of glioma on stroke is complex; therefore, each phase should be studied separately. In the context of glioma, ischemic stroke treatment also faces challenges, particularly in the titration of anticoagulant or antiplatelet medications.

Though the therapeutic potential of glioma-derived EVs appears promising, the therapeutic application of glioma-derived EVs remains challenging and requires further investigation. Firstly, as EVs serve as therapeutic carriers, delivery efficiency and potential off-target effects must be carefully evaluated, particularly in the context of the blood-brain barrier [249]. Secondly, given that EVs are biologically isolated vesicles, their heterogeneity poses a major challenge in ensuring reproducibility and therapeutic consistency [250, 251]. Thirdly, considering the distinct pathophysiological stages of ischemic stroke, optimal administration timing is a crucial factor that must be optimized to maximize therapeutic efficacy. Further research is warranted in the areas of drug delivery strategies, precise administration timing, advanced purification techniques, and scalable biomanufacturing processes to enhance the clinical translation of glioma-derived EVs as a viable therapeutic approach.

Conclusion

The pathology of glioma and ischemic stroke significantly overlaps in their pathological processes, including the regulation of cellular metabolism, inflammation, coagulation, hypoxia, angiogenesis, and neural repair, all of which involve shared biological factors. This overlap is reflected in numerous clinical studies. Glioma-derived EVs play a crucial role in mediating intracranial interactions and shaping the tumor microenvironment, with their cargos closely aligning with stroke-related biological factors, which act as risk factors and participating in the phases of hypoxia, stroke onset, progression, and recovery. Therefore, though further studies are needed to confirm their precise role, EVs have the potential to mediate interactions between glioma and stroke and suggest therapeutic potential.

Data availability

No datasets were generated or analysed during the current study.

References

  1. Schaff LR, Mellinghoff IK. Glioblastoma and other primary brain malignancies in adults: A review. JAMA. 2023;329(7):574–87.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Weller M, et al. Glioma Nat Rev Dis Primers. 2015;1:15017.

  3. Saini V, Guada L, Yavagal DR. Global epidemiology of stroke and access to acute ischemic stroke interventions. Neurology. 2021;97(20 Suppl 2):S6–16.

    PubMed  Google Scholar 

  4. Lee EC et al. Utility of exosomes in ischemic and hemorrhagic stroke diagnosis and treatment. Int J Mol Sci, 2022. 23(15).

  5. Gempt J, et al. Postoperative ischemic changes following resection of newly diagnosed and recurrent gliomas and their clinical relevance. J Neurosurg. 2013;118(4):801–8.

    Article  PubMed  Google Scholar 

  6. Talos I-F, et al. Supratentorial low-grade glioma resectability: statistical predictive analysis based on anatomic MR features and tumor characteristics. Radiology. 2006;239(2):506–13.

    Article  PubMed  Google Scholar 

  7. Ghosh MK, et al. The interrelationship between cerebral ischemic stroke and glioma: a comprehensive study of recent reports. Signal Transduct Target Ther. 2019;4:42.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Bond LM, Skrobo D. Multiple embolic cerebral infarcts as the first manifestation of metastatic ovarian cancer. BMJ Case Rep. 2015;2015.

  9. Kreisl TN, et al. Ischemic stroke in patients with primary brain tumors. Neurology. 2008;70(24):2314–20.

    Article  PubMed  Google Scholar 

  10. Bitzer M, Topka H. Progressive cerebral occlusive disease after radiation therapy. Stroke. 1995;26(1):131–6.

    Article  CAS  PubMed  Google Scholar 

  11. Qureshi AI, et al. Incident cancer in a cohort of 3,247 cancer diagnosis free ischemic stroke patients. Cerebrovasc Dis. 2015;39(5–6):262–8.

    Article  PubMed  Google Scholar 

  12. Wojtasiewicz TJ et al. De novo glioblastoma in the territory of a prior middle cerebral artery infarct. Case Rep Neurol Med. 2013;2013:356526.

  13. Lan Z, Li X, Zhang X. Glioblastoma: an update in pathology, molecular mechanisms and biomarkers. Int J Mol Sci, 2024. 25(5).

  14. Fan PL, et al. Time-dependent dual effect of microglia in ischemic stroke. Neurochem Int. 2023;169:105584.

    Article  CAS  PubMed  Google Scholar 

  15. Dornak T, et al. Posterior vs. anterior circulation infarction: demography, outcomes, and frequency of hemorrhage after thrombolysis. Int J Stroke. 2015;10(8):1224–8.

    Article  PubMed  Google Scholar 

  16. Fang J, Wang Z, Miao CY. Angiogenesis after ischemic stroke. Acta Pharmacol Sin. 2023;44(7):1305–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Guo X, Sui R, Piao H. Exosomes-mediated crosstalk between glioma and immune cells in the tumor microenvironment. CNS Neurosci Ther. 2023;29(8):2074–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Radin DP, Tsirka SE. Interactions between tumor cells, neurons, and microglia in the glioma microenvironment. Int J Mol Sci, 2020. 21(22).

  19. Kwak SB, et al. Tumor regionalization after surgery: roles of the tumor microenvironment and neutrophil extracellular traps. Exp Mol Med. 2022;54(6):720–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yekula A, et al. Extracellular vesicles in glioblastoma tumor microenvironment. Frontiers in Immunology; 2020. p. 10.

  21. Liu H, et al. Therapeutic prospects of MicroRNAs carried by mesenchymal stem cells-derived extracellular vesicles in autoimmune diseases. Life Sci. 2021;277:119458.

    Article  CAS  PubMed  Google Scholar 

  22. Balakrishnan A et al. The emerging role of extracellular vesicles in the glioma microenvironment: biogenesis and clinical relevance. Cancers (Basel), 2020. 12(7).

  23. Cela I, et al. Extracellular vesicles in glioblastoma: biomarkers and therapeutic tools. Semin Cancer Biol. 2024;101:25–43.

    Article  CAS  PubMed  Google Scholar 

  24. Chistiakov DA, Chekhonin VP. Extracellular vesicles shed by glioma cells: pathogenic role and clinical value. Tumour Biol. 2014;35(9):8425–38.

    Article  CAS  PubMed  Google Scholar 

  25. Mondal A, et al. Extracellular vesicles as modulators of tumor microenvironment and disease progression in glioma. Front Oncol. 2017;7:144.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Schulz-Siegmund M, Aigner A. Nucleic acid delivery with extracellular vesicles. Adv Drug Deliv Rev. 2021;173:89–111.

    Article  CAS  PubMed  Google Scholar 

  27. Nieland L, et al. Engineered EVs designed to target diseases of the CNS. J Control Release. 2023;356:493–506.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lotvall J, et al. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the international society for extracellular vesicles. J Extracell Vesicles. 2014;3:26913.

    Article  PubMed  Google Scholar 

  29. Couch Y, et al. A brief history of nearly EV-erything - The rise and rise of extracellular vesicles. J Extracell Vesicles. 2021;10(14):e12144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Jeppesen DK, et al. Reassessment of exosome composition. Cell. 2019;177(2):428–e44518.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Minciacchi VR, Freeman MR, Di Vizio D. Extracellular vesicles in cancer: exosomes, microvesicles and the emerging role of large oncosomes. Semin Cell Dev Biol. 2015;40:41–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dixson AC, et al. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat Rev Mol Cell Biol. 2023;24(7):454–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Pulliam L, et al. Plasma neuronal exosomes serve as biomarkers of cognitive impairment in HIV infection and Alzheimer’s disease. J Neurovirol. 2019;25(5):702–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lasser C, et al. Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med. 2011;9:9.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Welton JL, et al. Cerebrospinal fluid extracellular vesicle enrichment for protein biomarker discovery in neurological disease; multiple sclerosis. J Extracell Vesicles. 2017;6(1):1369805.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Mirza AH, et al. Breast Milk-Derived extracellular vesicles enriched in exosomes from mothers with type 1 diabetes contain aberrant levels of MicroRNAs. Front Immunol. 2019;10:2543.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Street JM, et al. Urine exosomes: an emerging trove of biomarkers. Adv Clin Chem. 2017;78:103–22.

    Article  CAS  PubMed  Google Scholar 

  38. Vojtech L, et al. Exosomes in human semen carry a distinctive repertoire of small non-coding RNAs with potential regulatory functions. Nucleic Acids Res. 2014;42(11):7290–304.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Peinado H, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 2012;18(6):883–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Singh AB, Harris RC. Autocrine, paracrine and juxtacrine signaling by EGFR ligands. Cell Signal. 2005;17(10):1183–93.

    Article  CAS  PubMed  Google Scholar 

  41. Yanez-Mo M, et al. Biological properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015;4:27066.

    Article  PubMed  Google Scholar 

  42. Thery C, et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the international society for extracellular vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7(1):1535750.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Pan Y, et al. Extracellular vesicles as delivery shippers for noncoding RNA-Based modulation of angiogenesis: insights from ischemic stroke and Cancer. Small. 2023;19(17):e2205739.

    Article  PubMed  Google Scholar 

  44. Anastasi F, et al. Proteomics analysis of Circulating small extracellular vesicles: focus on the contribution of EVs to tumor metabolism. Cytokine Growth Factor Rev. 2023;73:3–19.

    Article  CAS  PubMed  Google Scholar 

  45. GRAUS F, ROGERS LR, POSNER JB. Cerebrovascular complications in patients with cancer. Medicine. 1985;64(1):16–35.

    Article  CAS  PubMed  Google Scholar 

  46. Grisold W, Oberndorfer S, Struhal W. Stroke and cancer: a review. Acta Neurol Scand. 2009;119(1):1–16.

    Article  CAS  PubMed  Google Scholar 

  47. Schlehofer B, et al. Medical risk factors and the development of brain tumors. Cancer. 1992;69(10):2541–7.

    Article  CAS  PubMed  Google Scholar 

  48. Chen CW, et al. Increased risk of brain cancer incidence in stroke patients: a clinical case series, population-based and longitudinal follow-up study. Oncotarget. 2017;8(65):108989–99.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Cote DJ, Dawood HY, Smith TR. Venous thromboembolism In patients with high-grade glioma. In Seminars In thrombosis and hemostasis. Thieme Medical; 2016.

  50. Perry JR. Thromboembolic disease in patients with high-grade glioma. Neurooncology. 2012;14(suppl4):iv73–80.

    CAS  Google Scholar 

  51. Ivan ME, et al. Epidermal growth factor-like module containing mucin-like hormone receptor 2 expression in gliomas. J Neurooncol. 2015;121:53–61.

    Article  CAS  PubMed  Google Scholar 

  52. Riedl J, et al. Podoplanin expression in primary brain tumors induces platelet aggregation and increases risk of venous thromboembolism. Blood J Am Soc Hematol. 2017;129(13):1831–9.

    CAS  Google Scholar 

  53. Demers M, Wagner DD. NETosis: a new factor in tumor progression and cancer-associated thrombosis. Seminars in thrombosis and hemostasis. Thieme Medical; 2014.

  54. Rinaldi M et al. ROS and brain gliomas: an overview of potential and innovative therapeutic strategies. Int J Mol Sci, 2016. 17(6).

  55. Liou GY, Storz P. Reactive oxygen species in cancer. Free Radic Res. 2010;44(5):479–96.

    Article  CAS  PubMed  Google Scholar 

  56. Olmez I, Ozyurt H. Reactive oxygen species and ischemic cerebrovascular disease. Neurochem Int. 2012;60(2):208–12.

    Article  CAS  PubMed  Google Scholar 

  57. Wang YF, Hu JY. Natural and synthetic compounds for glioma treatment based on ROS-mediated strategy. Eur J Pharmacol. 2023;953:175537.

    Article  CAS  PubMed  Google Scholar 

  58. Obermeier B, Daneman R, Ransohoff RM. Development, maintenance and disruption of the blood-brain barrier. Nat Med. 2013;19(12):1584–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Noell S, et al. An allograft glioma model reveals the dependence of aquaporin-4 expression on the brain microenvironment. PLoS ONE. 2012;7(5):e36555.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Wang Y, et al. Remodelling and treatment of the Blood-Brain barrier in glioma. Cancer Manag Res. 2021;13:4217–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Zhao C, et al. Hypoxic glioblastoma release Exosomal VEGF-A induce the permeability of blood-brain barrier. Biochem Biophys Res Commun. 2018;502(3):324–31.

    Article  CAS  PubMed  Google Scholar 

  62. Fecci PE, Sampson JH. The current state of immunotherapy for gliomas: an eye toward the future. J Neurosurg. 2019;131(3):657–66.

    Article  PubMed  Google Scholar 

  63. Keaney J, Campbell M. The dynamic blood–brain barrier. FEBS J. 2015;282(21):4067–79.

    Article  CAS  PubMed  Google Scholar 

  64. Jiang X, et al. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol. 2018;163–164:144–71.

    Article  PubMed  Google Scholar 

  65. Rosenberg GA. Ischemic brain edema. Prog Cardiovasc Dis. 1999;42(3):209–16.

    Article  CAS  PubMed  Google Scholar 

  66. Huang J, Upadhyay UM, Tamargo RJ. Inflammation in stroke and focal cerebral ischemia. Surg Neurol. 2006;66(3):232–45.

    Article  PubMed  Google Scholar 

  67. Sweeney MD, Ayyadurai S, Zlokovic BV. Pericytes of the neurovascular unit: key functions and signaling pathways. Nat Neurosci. 2016;19(6):771–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zhao Z, et al. Establishment and dysfunction of the Blood-Brain barrier. Cell. 2015;163(5):1064–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hall CN, et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature. 2014;508(7494):55–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Dalkara T, Alarcon-Martinez L, Yemisci M. Pericytes in ischemic stroke. Adv Exp Med Biol. 2019;1147:189–213.

    Article  CAS  PubMed  Google Scholar 

  71. Cao L, et al. Pericytes for therapeutic approaches to ischemic stroke. Front Neurosci. 2021;15:629297.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Valdor R, et al. Glioblastoma progression is assisted by induction of immunosuppressive function of pericytes through interaction with tumor cells. Oncotarget. 2017;8(40):68614.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Garcia C, et al. The orthotopic xenotransplant of human glioblastoma successfully recapitulates glioblastoma-microenvironment interactions in a non-immunosuppressed mouse model. BMC Cancer. 2014;14(1):1–11.

    Article  Google Scholar 

  74. Huizer K, et al. Periostin is expressed by pericytes and is crucial for angiogenesis in glioma. J Neuropathology Experimental Neurol. 2020;79(8):863–72.

    Article  CAS  Google Scholar 

  75. Ahsan MS, et al. Differential expression of Perlecan receptors, α-dystroglycan and integrin Β1, before and after invasion of oral squamous cell carcinoma. J Oral Pathol Med. 2011;40(7):552–9.

    Article  CAS  PubMed  Google Scholar 

  76. Hu X et al. Role of Glial Cell-Derived Oxidative Stress in Blood-Brain Barrier Damage after Acute Ischemic Stroke. Oxid Med Cell Longev. 2022;2022:7762078.

  77. George EL, et al. Defects in mesoderm, neural tube and vascular development in mouse embryos lacking fibronectin. Development. 1993;119(4):1079–91.

    Article  CAS  PubMed  Google Scholar 

  78. Shen W, et al. Tyrosine phosphorylation of VE-cadherin and claudin-5 is associated with TGF-β1-induced permeability of centrally derived vascular endothelium. Eur J Cell Biol. 2011;90(4):323–32.

    Article  CAS  PubMed  Google Scholar 

  79. Montana V, Sontheimer H. Bradykinin promotes the chemotactic invasion of primary brain tumors. J Neurosci. 2011;31(13):4858–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Zagzag D, et al. Vascular apoptosis and Involution in gliomas precede neovascularization: a novel concept for glioma growth and angiogenesis. Lab Invest. 2000;80(6):837–49.

    Article  CAS  PubMed  Google Scholar 

  81. Watkins S, et al. Disruption of astrocyte–vascular coupling and the blood–brain barrier by invading glioma cells. Nat Commun. 2014;5(1):4196.

    Article  CAS  PubMed  Google Scholar 

  82. Le DM, et al. Exploitation of astrocytes by glioma cells to facilitate invasiveness: a mechanism involving matrix metalloproteinase-2 and the urokinase-type plasminogen activator–plasmin cascade. J Neurosci. 2003;23(10):4034–43.

    Article  PubMed  PubMed Central  Google Scholar 

  83. Tanabe S et al. Functional expression of the CXC-chemokine receptor-4/fusin on mouse microglial cells and astrocytes. Journal of immunology (Baltimore, Md.: 1950). 1997;159(2):905–911.

  84. Zhang L, Zhang Y. Tunneling nanotubes between rat primary astrocytes and C6 glioma cells alter proliferation potential of glioma cells. Neurosci Bull. 2015;31:371–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Shabtay-Orbach A, et al. Paracrine regulation of glioma cells invasion by astrocytes is mediated by glial‐derived neurotrophic factor. Int J Cancer. 2015;137(5):1012–20.

    Article  CAS  PubMed  Google Scholar 

  86. Xin WQ, et al. Modulating poststroke inflammatory mechanisms: novel aspects of mesenchymal stem cells, extracellular vesicles and microglia. World J Stem Cells. 2021;13(8):1030–48.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Hu X, et al. Microglial and macrophage polarization—new prospects for brain repair. Nat Reviews Neurol. 2015;11(1):56–64.

    Article  Google Scholar 

  88. Zhang L, Zhang J, You Z. Switching of the microglial activation phenotype is a possible treatment for depression disorder. Front Cell Neurosci. 2018;12:306.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Ma L, et al. PGC-1α-mediated mitochondrial biogenesis is involved in cannabinoid receptor 2 agonist AM1241-induced microglial phenotype amelioration. Cell Mol Neurobiol. 2018;38:1529–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Zeng J, et al. The mechanism of microglia-mediated immune inflammation in ischemic stroke and the role of natural botanical components in regulating microglia: A review. Front Immunol. 2022;13:1047550.

    Article  CAS  PubMed  Google Scholar 

  91. Graeber MB, Scheithauer BW, Kreutzberg GW. Microglia in brain tumors. Glia. 2002;40(2):252–9.

    Article  PubMed  Google Scholar 

  92. Li W, Graeber MB. The molecular profile of microglia under the influence of glioma. Neuro Oncol. 2012;14(8):958–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Schaeffer S, Iadecola C. Revisiting the neurovascular unit. Nat Neurosci. 2021;24(9):1198–209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Kretschmer M, Rüdiger D, Zahler S. Mechanical Aspects of Angiogenesis. Cancers (Basel). 2021;13(19).

  95. Jiang X, et al. The role of microenvironment in tumor angiogenesis. J Experimental Clin cancer Research: CR. 2020;39(1):204.

    Article  PubMed Central  Google Scholar 

  96. Hatakeyama M, Ninomiya I, Kanazawa M. Angiogenesis and neuronal remodeling after ischemic stroke. Neural Regen Res. 2020;15(1):16–9.

    Article  PubMed  Google Scholar 

  97. Liao D, Johnson R. Hypoxia: a key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007;26(2):281–90.

    Article  CAS  PubMed  Google Scholar 

  98. Sun Z, et al. Emerging role of exosome-derived long non-coding RNAs in tumor microenvironment. Mol Cancer. 2018;17(1):82.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Parkin J, Cohen B. An overview of the immune system. Lancet. 2001;357(9270):1777–89.

    Article  CAS  PubMed  Google Scholar 

  100. Hu X, et al. Microglial and macrophage polarization-new prospects for brain repair. Nat Rev Neurol. 2015;11(1):56–64.

    Article  PubMed  Google Scholar 

  101. Becker KJ. Modulation of the postischemic immune response to improve stroke outcome. Stroke. 2010;41(10 Suppl):S75–8.

    PubMed  PubMed Central  Google Scholar 

  102. Dougan M, Dranoff G, Dougan SK. GM-CSF, IL-3, and IL-5 family of cytokines: regulators of inflammation. Immunity. 2019;50(4):796–811.

    Article  CAS  PubMed  Google Scholar 

  103. Pyonteck SM, et al. CSF-1R Inhibition alters macrophage polarization and blocks glioma progression. Nat Med. 2013;19(10):1264–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Abdelfattah N, et al. Single-cell analysis of human glioma and immune cells identifies S100A4 as an immunotherapy target. Nat Commun. 2022;13(1):767.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Chen H, et al. Immune response in glioma’s microenvironment. Innov Surg Sci. 2020;5(3–4):20190001.

    PubMed  Google Scholar 

  106. Zisakis A, et al. Comparative analysis of peripheral and localised cytokine secretion in glioblastoma patients. Cytokine. 2007;39(2):99–105.

    Article  CAS  PubMed  Google Scholar 

  107. Matias D, et al. Microglia/astrocytes–glioblastoma crosstalk: crucial molecular mechanisms and microenvironmental factors. Front Cell Neurosci. 2018;12:235.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Yang Z, et al. The role and application of small extracellular vesicles in glioma. Cancer Cell Int. 2024;24(1):229.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Guescini M, et al. Astrocytes and glioblastoma cells release exosomes carrying MtDNA. J Neural Transm (Vienna). 2010;117(1):1–4.

    Article  CAS  PubMed  Google Scholar 

  110. Shao H, et al. Protein typing of Circulating microvesicles allows real-time monitoring of glioblastoma therapy. Nat Med. 2012;18(12):1835–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Cumba Garcia LM, et al. Isolation and analysis of Plasma-Derived exosomes in patients with glioma. Front Oncol. 2019;9:651.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Pace KR, Dutt R, Galileo DS. Exosomal L1CAM stimulates glioblastoma cell motility, proliferation, and invasiveness. Int J Mol Sci, 2019. 20(16).

  113. Takacs GP et al. Glioma-derived CCL2 and CCL7 mediate migration of immune suppressive CCR2+/CX3CR1 + M-MDSCs into the tumor microenvironment in a redundant manner. Front Immunol, 2023. 13.

  114. van der Vos KE, et al. Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 2016;18(1):58–69.

    Article  PubMed  Google Scholar 

  115. Graner MW, et al. Proteomic and Immunologic analyses of brain tumor exosomes. FASEB J. 2009;23(5):1541–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Ravi VM et al. T-cell dysfunction in the glioblastoma microenvironment is mediated by myeloid cells releasing interleukin-10. Nat Commun, 2022. 13(1).

  117. Alban TJ et al. Global immune fingerprinting in glioblastoma patient peripheral blood reveals immune-suppression signatures associated with prognosis. JCI Insight, 2018. 3(21).

  118. Domenis R, et al. Systemic T cells immunosuppression of glioma stem Cell-Derived exosomes is mediated by monocytic Myeloid-Derived suppressor cells. PLoS ONE. 2017;12(1):e0169932.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Guo X, et al. Immunosuppressive effects of hypoxia-induced glioma exosomes through myeloid-derived suppressor cells via the miR-10a/Rora and miR-21/Pten pathways. Oncogene. 2018;37(31):4239–59.

    Article  CAS  PubMed  Google Scholar 

  120. Shah D, et al. A novel miR1983-TLR7-IFNbeta circuit licenses NK cells to kill glioma cells, and is under the control of galectin-1. Oncoimmunology. 2021;10(1):1939601.

    Article  PubMed  PubMed Central  Google Scholar 

  121. Liu ZM, Wang YB, Yuan XH. Exosomes from murine-derived GL26 cells promote glioblastoma tumor growth by reducing number and function of CD8 + T cells. Asian Pac J Cancer Prev. 2013;14(1):309–14.

    Article  PubMed  Google Scholar 

  122. Yang JK, et al. Exosomal miR-214-5p released from glioblastoma cells modulates inflammatory response of microglia after lipopolysaccharide stimulation through targeting CXCR5. CNS Neurol Disord Drug Targets. 2019;18(1):78–87.

    Article  CAS  PubMed  Google Scholar 

  123. Qian M, et al. Hypoxic glioma-derived exosomes deliver microRNA-1246 to induce M2 macrophage polarization by targeting TERF2IP via the STAT3 and NF-kappaB pathways. Oncogene. 2020;39(2):428–42.

    Article  CAS  PubMed  Google Scholar 

  124. Mongiardi MP. Angiogenesis and hypoxia in glioblastoma: a focus on cancer stem cells. CNS Neurol Disord Drug Targets. 2012;11(7):878–83.

    Article  CAS  PubMed  Google Scholar 

  125. Lang HL, et al. Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2. Oncol Rep. 2017;38(2):785–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Wang ZF, et al. Glioma stem cells-derived Exosomal miR-26a promotes angiogenesis of microvessel endothelial cells in glioma. J Exp Clin Cancer Res. 2019;38(1):201.

    Article  PubMed  PubMed Central  Google Scholar 

  127. Skog J, et al. Glioblastoma microvesicles transport RNA and proteins that promote tumour growth and provide diagnostic biomarkers. Nat Cell Biol. 2008;10(12):1470–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Li CC, et al. Glioma microvesicles carry selectively packaged coding and non-coding RNAs which alter gene expression in recipient cells. RNA Biol. 2013;10(8):1333–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Kucharzewska P, et al. Exosomes reflect the hypoxic status of glioma cells and mediate hypoxia-dependent activation of vascular cells during tumor development. Proc Natl Acad Sci U S A. 2013;110(18):7312–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Lang HL, et al. Glioma cells promote angiogenesis through the release of exosomes containing long non-coding RNA POU3F3. Eur Rev Med Pharmacol Sci. 2017;21(5):959–72.

    PubMed  Google Scholar 

  131. Bronisz A, et al. Extracellular vesicles modulate the glioblastoma microenvironment via a tumor suppression signaling network directed by miR-1. Cancer Res. 2014;74(3):738–50.

    Article  CAS  PubMed  Google Scholar 

  132. Sun X, et al. Glioma stem cells-derived exosomes promote the angiogenic ability of endothelial cells through miR-21/VEGF signal. Oncotarget. 2017;8(22):36137–48.

    Article  PubMed  PubMed Central  Google Scholar 

  133. Akers JC, et al. MiR-21 in the extracellular vesicles (EVs) of cerebrospinal fluid (CSF): a platform for glioblastoma biomarker development. PLoS ONE. 2013;8(10):e78115.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  134. Figueroa JM, et al. Detection of wild-type EGFR amplification and EGFRvIII mutation in CSF-derived extracellular vesicles of glioblastoma patients. Neuro Oncol. 2017;19(11):1494–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Kolominsky-Rabas PL, et al. Epidemiology of ischemic stroke subtypes according to TOAST criteria: incidence, recurrence, and long-term survival in ischemic stroke subtypes: a population-based study. Stroke. 2001;32(12):2735–40.

    Article  CAS  PubMed  Google Scholar 

  136. Chen PH, et al. Classifying ischemic stroke, from TOAST to CISS. CNS Neurosci Ther. 2012;18(6):452–6.

    Article  PubMed  PubMed Central  Google Scholar 

  137. Ohashi SN, et al. Role of inflammatory processes in hemorrhagic stroke. Stroke. 2023;54(2):605–19.

    Article  PubMed  Google Scholar 

  138. Broekman ML, et al. Multidimensional communication in the microenvirons of glioblastoma. Nat Rev Neurol. 2018;14(8):482–95.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Manwani B, et al. Small RNA signatures of acute ischemic stroke in L1CAM positive extracellular vesicles. Sci Rep. 2024;14(1):13560.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Colombo E, et al. Astrocyte TrkB promotes brain injury and edema formation in ischemic stroke. Neurobiol Dis. 2024;201:106670.

    Article  CAS  PubMed  Google Scholar 

  141. Pinet S, et al. TrkB-containing exosomes promote the transfer of glioblastoma aggressiveness to YKL-40-inactivated glioblastoma cells. Oncotarget. 2016;7(31):50349–64.

    Article  PubMed  PubMed Central  Google Scholar 

  142. Simone L, et al. AQP4-dependent glioma cell features affect the phenotype of surrounding cells via extracellular vesicles. Cell Biosci. 2022;12(1):150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Dai X, et al. AHIF promotes glioblastoma progression and radioresistance via exosomes. Int J Oncol. 2019;54(1):261–70.

    CAS  PubMed  Google Scholar 

  144. Longo V, et al. Transcranial direct current stimulation enhances neuroplasticity and accelerates motor recovery in a stroke mouse model. Stroke. 2022;53(5):1746–58.

    Article  CAS  PubMed  Google Scholar 

  145. Wang J, et al. Administration of intramuscular AAV-BDNF and intranasal AAV-TrkB promotes neurological recovery via enhancing corticospinal synaptic connections in stroke rats. Exp Neurol. 2023;359:114236.

    Article  CAS  PubMed  Google Scholar 

  146. Kang Z, et al. Effect of electroacupuncture at Siguan acupoints on expression of BDNF and TrkB proteins in the Hippocampus of Post-Stroke depression rats. J Mol Neurosci. 2021;71(10):2165–71.

    Article  CAS  PubMed  Google Scholar 

  147. Xu X, et al. Selective exosome exclusion of miR-375 by glioma cells promotes glioma progression by activating the CTGF-EGFR pathway. J Exp Clin Cancer Res. 2021;40(1):16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Salman AT, et al. The expression profiling of serum miR-92a, miR-134 and miR-375 in acute ischemic stroke. Future Sci OA. 2022;8(10):pFso829.

    Article  Google Scholar 

  149. Clement T, Rodriguez-Grande B, Badaut J. Aquaporins in brain edema. J Neurosci Res. 2020;98(1):9–18.

    Article  CAS  PubMed  Google Scholar 

  150. Gu Y, et al. Cerebral edema after ischemic stroke: pathophysiology and underlying mechanisms. Front Neurosci. 2022;16:988283.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Cai Q, Zhu A, Gong L. Exosomes of glioma cells deliver miR-148a to promote proliferation and metastasis of glioblastoma via targeting CADM1. Bull Cancer. 2018;105(7–8):643–51.

    Article  PubMed  Google Scholar 

  152. Jickling GC, et al. MicroRNA expression in peripheral blood cells following acute ischemic stroke and their predicted gene targets. PLoS ONE. 2014;9(6):e99283.

    Article  PubMed  PubMed Central  Google Scholar 

  153. Zhao X, et al. Machine learning analysis of MicroRNA expression data reveals novel diagnostic biomarker for ischemic stroke. J Stroke Cerebrovasc Dis. 2021;30(8):105825.

    Article  PubMed  Google Scholar 

  154. Zhao M, et al. Expression profiles and potential functions of circular RNAs in extracellular vesicles isolated from radioresistant glioma cells. Oncol Rep. 2019;41(3):1893–900.

    CAS  PubMed  Google Scholar 

  155. Luo D et al. Regulation of a novel circATP8B4/miR-31-5p/nestin CeRNA crosstalk in proliferation, motility, invasion and radiosensitivity of human glioma cells. J Radiat Res, 2024.

  156. Yang JK, et al. Exosomal miR-221 targets DNM3 to induce tumor progression and Temozolomide resistance in glioma. J Neurooncol. 2017;131(2):255–65.

    Article  CAS  PubMed  Google Scholar 

  157. Zeng A, et al. Exosomal transfer of miR-151a enhances chemosensitivity to Temozolomide in drug-resistant glioblastoma. Cancer Lett. 2018;436:10–21.

    Article  CAS  PubMed  Google Scholar 

  158. Yin J, et al. Extracellular vesicles derived from hypoxic glioma stem-like cells confer Temozolomide resistance on glioblastoma by delivering miR-30b-3p. Theranostics. 2021;11(4):1763–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Shan Y, et al. miR-221 exerts neuroprotective effects in ischemic stroke by inhibiting the Proinflammatory response. J Stroke Cerebrovasc Dis. 2021;30(2):105489.

    Article  PubMed  Google Scholar 

  160. Chang H, Lu Z. The expression of serummiR-151a-3p in patients with acute cerebral infarction and its correlation with pro-inflammatory factors]. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue. 2016;28(3):272–6.

    PubMed  Google Scholar 

  161. He Q, et al. Biological functions and regulatory mechanisms of Hypoxia-Inducible Factor-1alpha in ischemic stroke. Front Immunol. 2021;12:801985.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Mu D, et al. Regulation of hypoxia-inducible factor 1alpha and induction of vascular endothelial growth factor in a rat neonatal stroke model. Neurobiol Dis. 2003;14(3):524–34.

    Article  CAS  PubMed  Google Scholar 

  163. Sharp FR, Bergeron M, Bernaudin M. Hypoxia-inducible factor in brain. Adv Exp Med Biol. 2001;502:273–91.

    Article  CAS  PubMed  Google Scholar 

  164. Cui Y, et al. ACSL4 exacerbates ischemic stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav Immun. 2021;93:312–21.

    Article  CAS  PubMed  Google Scholar 

  165. Li HS, et al. HIF-1alpha protects against oxidative stress by directly targeting mitochondria. Redox Biol. 2019;25:101109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Slevin M, et al. Can angiogenesis be exploited to improve stroke outcome? Mechanisms and therapeutic potential. Clin Sci (Lond). 2006;111(3):171–83.

    Article  CAS  PubMed  Google Scholar 

  167. Mao L, et al. Endogenous endothelial progenitor cells participate in neovascularization via CXCR4/SDF-1 axis and improve outcome after stroke. CNS Neurosci Ther. 2014;20(5):460–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Chopp M, Zhang ZG, Jiang Q. Neurogenesis, angiogenesis, and MRI indices of functional recovery from stroke. Stroke. 2007;38(2 Suppl):827–31.

    Article  PubMed  Google Scholar 

  169. Wurmser AE, Palmer TD, Gage FH. Neurosci Cell Interact Stem Cell Niche Sci. 2004;304(5675):1253–5.

  170. LaManna JC, Chavez JC, Pichiule P. Structural and functional adaptation to hypoxia in the rat brain. J Exp Biol. 2004;207(18):3163–9.

    Article  CAS  PubMed  Google Scholar 

  171. Halder SK, Milner R. The impact of chronic mild hypoxia on cerebrovascular remodelling; uncoupling of angiogenesis and vascular breakdown. Fluids Barriers CNS. 2021;18(1):50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Burke GM, et al. Moyamoya disease: a summary. Neurosurg Focus. 2009;26(4):E11.

    Article  PubMed  Google Scholar 

  173. Shi W, et al. Hypoxic postconditioning promotes angiogenesis after ischemic stroke. Neuroscience. 2023;526:35–47.

    Article  CAS  PubMed  Google Scholar 

  174. Giusti I, et al. From glioblastoma to endothelial cells through extracellular vesicles: messages for angiogenesis. Tumour Biol. 2016;37(9):12743–53.

    Article  CAS  PubMed  Google Scholar 

  175. Zorio E, et al. Fibrinolysis: the key to new pathogenetic mechanisms. Curr Med Chem. 2008;15(9):923–9.

    Article  CAS  PubMed  Google Scholar 

  176. Hajjar KA, Deora A. New concepts in fibrinolysis and angiogenesis. Curr Atheroscler Rep. 2000;2(5):417–21.

    Article  CAS  PubMed  Google Scholar 

  177. Nguyen JN, et al. CD13 facilitates immune cell migration and aggravates acute injury but promotes chronic post-stroke recovery. J Neuroinflammation. 2023;20(1):232.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Zhang MF, et al. Catalpol attenuates ischemic stroke by promoting neurogenesis and angiogenesis via the SDF-1alpha/CXCR4 pathway. Phytomedicine. 2024;128:155362.

    Article  CAS  PubMed  Google Scholar 

  179. Jiang G, et al. Remote ischemic postconditioning ameliorates stroke injury via the SDF-1α/CXCR4 signaling axis in rats. Brain Res Bull. 2023;197:31–41.

    Article  CAS  PubMed  Google Scholar 

  180. Bang OY, et al. Stroke induces mesenchymal stem cell migration to infarcted brain areas via CXCR4 and C-Met signaling. Transl Stroke Res; 2017.

  181. Geiseler SJ, Morland C. The Janus face of VEGF in stroke. Int J Mol Sci, 2018. 19(5).

  182. Ma Y, et al. Effects of vascular endothelial growth factor in ischemic stroke. J Neurosci Res. 2012;90(10):1873–82.

    Article  CAS  PubMed  Google Scholar 

  183. Kurzepa J, et al. The significance of matrix metalloproteinase (MMP)-2 and MMP-9 in the ischemic stroke. Int J Neurosci. 2014;124(10):707–16.

    Article  CAS  PubMed  Google Scholar 

  184. Cai H, et al. Hypoxia response Element-Regulated MMP-9 promotes neurological recovery via glial Scar degradation and angiogenesis in delayed stroke. Mol Ther. 2017;25(6):1448–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Zhang L, et al. Extracellular vesicles from hypoxia-preconditioned microglia promote angiogenesis and repress apoptosis in stroke mice via the TGF-beta/Smad2/3 pathway. Cell Death Dis. 2021;12(11):1068.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  186. Blochet C, et al. Involvement of caveolin-1 in neurovascular unit remodeling after stroke: effects on neovascularization and astrogliosis. J Cereb Blood Flow Metab. 2020;40(1):163–76.

    Article  CAS  PubMed  Google Scholar 

  187. Rajkovic I, et al. Pentraxin 3 promotes long-term cerebral blood flow recovery, angiogenesis, and neuronal survival after stroke. J Mol Med (Berl). 2018;96(12):1319–32.

    Article  CAS  PubMed  Google Scholar 

  188. Folestad E, Kunath A, Wagsater D. PDGF-C and PDGF-D signaling in vascular diseases and animal models. Mol Aspects Med. 2018;62:1–11.

    Article  CAS  PubMed  Google Scholar 

  189. Bernard M, et al. Endothelial PDGF-D contributes to neurovascular protection after ischemic stroke by rescuing pericyte functions. Cell Mol Life Sci. 2024;81(1):225.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Lin CM, et al. Ferulic acid augments angiogenesis via VEGF, PDGF and HIF-1 alpha. J Nutr Biochem. 2010;21(7):627–33.

    Article  CAS  PubMed  Google Scholar 

  191. Navarro-Sobrino M, et al. A large screening of angiogenesis biomarkers and their association with neurological outcome after ischemic stroke. Atherosclerosis. 2011;216(1):205–11.

    Article  CAS  PubMed  Google Scholar 

  192. Hou Y, et al. IL-8 enhances the angiogenic potential of human bone marrow mesenchymal stem cells by increasing vascular endothelial growth factor. Cell Biol Int. 2014;38(9):1050–9.

    Article  CAS  PubMed  Google Scholar 

  193. Xu J, et al. Hypoxic glioma-derived exosomes promote M2-like macrophage polarization by enhancing autophagy induction. Cell Death Dis. 2021;12(4):373.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Choi BR, et al. Monocyte-derived IL-6 programs microglia to rebuild damaged brain vasculature. Nat Immunol. 2023;24(7):1110–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Yin KJ, Hamblin M, Chen YE. Angiogenesis-regulating MicroRNAs and ischemic stroke. Curr Vasc Pharmacol. 2015;13(3):352–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Heydari E, et al. The role of non-coding RNAs in neuroprotection and angiogenesis following ischemic stroke. Metab Brain Dis. 2020;35(1):31–43.

    Article  CAS  PubMed  Google Scholar 

  197. E S, et al. The Circulating non-coding RNA landscape for biomarker research: lessons and prospects from cardiovascular diseases. Acta Pharmacol Sin. 2018;39(7):1085–99.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Ma X, et al. Long non-coding RNA HOTAIR enhances angiogenesis by induction of VEGFA expression in glioma cells and transmission to endothelial cells via glioma cell derived-extracellular vesicles. Am J Transl Res. 2017;9(11):5012–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Yin KJ, Hamblin M, Chen YE. Non-coding RNAs in cerebral endothelial pathophysiology: emerging roles in stroke. Neurochem Int. 2014;77:9–16.

    Article  CAS  PubMed  Google Scholar 

  200. Liang Z, et al. MiRNA-26a promotes angiogenesis in a rat model of cerebral infarction via PI3K/AKT and MAPK/ERK pathway. Eur Rev Med Pharmacol Sci. 2018;22(11):3485–92.

    CAS  PubMed  Google Scholar 

  201. Li F, et al. Taurine-Upregulated gene 1 attenuates cerebral angiogenesis following ischemic stroke in rats. Biomed Res Int. 2022;2022:p1037525.

    Article  Google Scholar 

  202. Zhang H, et al. Exosome-mediated targeted delivery of miR-210 for angiogenic therapy after cerebral ischemia in mice. J Nanobiotechnol. 2019;17(1):29.

    Article  Google Scholar 

  203. Wang Y, et al. LncRNA HOTAIR mediates OGD/R-induced cell injury and angiogenesis in a EZH2-dependent manner. Exp Ther Med. 2022;23(1):99.

    Article  PubMed  Google Scholar 

  204. Kyritsis N, et al. Acute inflammation initiates the regenerative response in the adult zebrafish brain. Science. 2012;338(6112):1353–6.

    Article  CAS  PubMed  Google Scholar 

  205. Planas AM. Role of immune cells migrating to the ischemic brain. Stroke. 2018;49(9):2261–7.

    Article  PubMed  Google Scholar 

  206. Zhang W, et al. Microglia-associated neuroinflammation is a potential therapeutic target for ischemic stroke. Neural Regen Res. 2021;16(1):6–11.

    Article  PubMed  Google Scholar 

  207. Wang M, et al. miR-451 protects against ischemic stroke by targeting Phd3. Exp Neurol. 2021;343:113777.

    Article  CAS  PubMed  Google Scholar 

  208. Chelluboina B, et al. Therapeutic potential of intravenous miR-21 mimic after stroke following STAIR criteria. Transl Stroke Res; 2023.

  209. Lopez MS, et al. MicroRNA miR-21 decreases Post-stroke brain damage in rodents. Transl Stroke Res. 2022;13(3):483–93.

    Article  CAS  PubMed  Google Scholar 

  210. Fu C, et al. Potential neuroprotective effect of miR-451 against cerebral ischemia/reperfusion injury in stroke patients and a mouse model. World Neurosurg. 2019;130:e54–61.

    Article  PubMed  Google Scholar 

  211. Li P, et al. Identification of Circulating MicroRNAs as potential biomarkers for detecting acute ischemic stroke. Cell Mol Neurobiol. 2015;35(3):433–47.

    Article  PubMed  Google Scholar 

  212. Zhu H, et al. Interleukins and ischemic stroke. Front Immunol. 2022;13:828447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Azambuja JH et al. Arginase-1 + Exosomes from reprogrammed macrophages promote glioblastoma progression. Int J Mol Sci, 2020. 21(11).

  214. Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32(1):19–25.

    Article  CAS  PubMed  Google Scholar 

  215. Yan H, et al. Role of polymorphonuclear Myeloid-Derived suppressor cells and neutrophils in ischemic stroke. J Am Heart Assoc. 2023;12(6):e028125.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  216. Zhang X, et al. BATF2 prevents glioblastoma multiforme progression by inhibiting recruitment of myeloid-derived suppressor cells. Oncogene. 2021;40(8):1516–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Hill WD, 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. 2004;63(1):84–96.

    Article  CAS  PubMed  Google Scholar 

  218. Malik M, et al. Monocyte migration and LFA-1-mediated attachment to brain microvascular endothelia is regulated by SDF-1 alpha through Lyn kinase. J Immunol. 2008;181(7):4632–7.

    Article  CAS  PubMed  Google Scholar 

  219. Qiu W, et al. Exosomal miR-1246 from glioma patient body fluids drives the differentiation and activation of myeloid-derived suppressor cells. Mol Ther. 2021;29(12):3449–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Qi Y, et al. The dual role of glioma Exosomal MicroRNAs: glioma eliminates tumor suppressor miR-1298-5p via exosomes to promote immunosuppressive effects of MDSCs. Cell Death Dis. 2022;13(5):426.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Zhao F, et al. MiR-29a knockout aggravates neurological damage by Pre-polarizing M1 microglia in experimental rat models of acute stroke. Front Genet. 2021;12:642079.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Zhao M, et al. Elevated miR-29a contributes to axonal outgrowth and neurological recovery after intracerebral hemorrhage via targeting PTEN/PI3K/Akt pathway. Cell Mol Neurobiol. 2021;41(8):1759–72.

    Article  CAS  PubMed  Google Scholar 

  223. Ouyang YB, et al. Astrocyte-enriched miR-29a targets PUMA and reduces neuronal vulnerability to forebrain ischemia. Glia. 2013;61(11):1784–94.

    Article  PubMed  Google Scholar 

  224. Lee DY, et al. Induction of microRNA-10a using retinoic acid receptor-alpha and retinoid X receptor-alpha agonists inhibits atherosclerotic lesion formation. Atherosclerosis. 2018;271:36–44.

    Article  CAS  PubMed  Google Scholar 

  225. Wang C, et al. Silencing of LncRNA XIST impairs angiogenesis and exacerbates cerebral vascular injury after ischemic stroke. Mol Ther Nucleic Acids. 2021;26:148–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Akers JC, et al. MiRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients. J Neurooncol. 2015;123(2):205–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  227. Santangelo A, et al. A MicroRNA signature from serum exosomes of patients with glioma as complementary diagnostic biomarker. J Neurooncol. 2018;136(1):51–62.

    Article  CAS  PubMed  Google Scholar 

  228. Shi R, et al. Exosomal levels of miRNA-21 from cerebrospinal fluids associated with poor prognosis and tumor recurrence of glioma patients. Oncotarget. 2015;6(29):26971–81.

    Article  PubMed  PubMed Central  Google Scholar 

  229. Buller B, et al. MicroRNA-21 protects neurons from ischemic death. FEBS J. 2010;277(20):4299–307.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  230. Yang Q, Yang K, Li A. microRNA-21 protects against ischemia-reperfusion and hypoxia-reperfusion-induced cardiocyte apoptosis via the phosphatase and tensin homolog/Akt-dependent mechanism. Mol Med Rep. 2014;9(6):2213–20.

    Article  CAS  PubMed  Google Scholar 

  231. Zhou J, Zhang J. Identification of miRNA-21 and miRNA-24 in plasma as potential early stage markers of acute cerebral infarction. Mol Med Rep. 2014;10(2):971–6.

    Article  CAS  PubMed  Google Scholar 

  232. Yuan M, et al. Diagnostic performance of miR-21, miR-124, miR-132, and miR-200b serums in post-stroke cognitive impairment patients. Folia Neuropathol. 2022;60(2):228–36.

    Article  PubMed  Google Scholar 

  233. Chernykh ER, et al. Safety and therapeutic potential of M2 macrophages in stroke treatment. Cell Transpl. 2016;25(8):1461–71.

    Article  Google Scholar 

  234. Coveney S, et al. Inflammatory cytokines, high-sensitivity C-reactive protein, and risk of one-year vascular events, death, and poor functional outcome after stroke and transient ischemic attack. Int J Stroke. 2022;17(2):163–71.

    Article  CAS  PubMed  Google Scholar 

  235. Lin SP, et al. Increased expression of microRNA-21 in peripheral blood mediates the down-regulation of IFN-gamma and increases the prevalence of stroke-associated infection. J Neurol Sci. 2016;366:235–9.

    Article  CAS  PubMed  Google Scholar 

  236. Klehmet J, et al. Stroke-induced immunodepression and post-stroke infections: lessons from the preventive antibacterial therapy in stroke trial. Neuroscience. 2009;158(3):1184–93.

    Article  CAS  PubMed  Google Scholar 

  237. Satani N, et al. A combination of Atorvastatin and aspirin enhances the Pro-Regenerative interactions of marrow stromal cells and Stroke-Derived monocytes in vitro. Front Pharmacol. 2021;12:589418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  238. Ricklefs FL, et al. Immune evasion mediated by PD-L1 on glioblastoma-derived extracellular vesicles. Sci Adv. 2018;4(3):eaar2766.

    Article  PubMed  PubMed Central  Google Scholar 

  239. Kim JE, et al. Soluble PD-L1 reprograms blood monocytes to prevent cerebral edema and facilitate recovery after ischemic stroke. Brain Behav Immun. 2024;116:160–74.

    Article  CAS  PubMed  Google Scholar 

  240. Li Y, et al. PD-L1 regulates platelet activation and thrombosis via Caspase-3/GSDME pathway. Front Pharmacol. 2022;13:921414.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  241. Bodhankar S, et al. PD-L1 monoclonal antibody treats ischemic stroke by controlling central nervous system inflammation. Stroke. 2015;46(10):2926–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Wang M, et al. CD73-positive extracellular vesicles promote glioblastoma immunosuppression by inhibiting T-cell clonal expansion. Cell Death Dis. 2021;12(11):1065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Petrovic-Djergovic D, et al. Tissue-resident ecto-5’ nucleotidase (CD73) regulates leukocyte trafficking in the ischemic brain. J Immunol. 2012;188(5):2387–98.

    Article  CAS  PubMed  Google Scholar 

  244. Schadlich IS, et al. Nt5e deficiency does not affect post-stroke inflammation and lesion size in a murine ischemia/reperfusion stroke model. iScience. 2022;25(6):104470.

    Article  PubMed  PubMed Central  Google Scholar 

  245. van Baarsen K, et al. Optic pathway-hypothalamic glioma hemorrhage: a series of 9 patients and review of the literature. J Neurosurg. 2018;129(6):1407–15.

    Article  PubMed  Google Scholar 

  246. Magnus N, et al. Brain neoplasms and coagulation. Semin Thromb Hemost. 2013;39(8):881–95.

    Article  CAS  PubMed  Google Scholar 

  247. Endres M, et al. Immune pathways in etiology, acute phase, and chronic sequelae of ischemic stroke. Circ Res. 2022;130(8):1167–86.

    Article  CAS  PubMed  Google Scholar 

  248. Yang Y, et al. M2 Macrophage-Derived exosomes promote angiogenesis and growth of pancreatic ductal adenocarcinoma by targeting E2F2. Mol Ther. 2021;29(3):1226–38.

    Article  CAS  PubMed  Google Scholar 

  249. Zhang W, et al. Remodeling brain pathological microenvironment to lessen cerebral ischemia injury by multifunctional injectable hydrogels. J Control Release. 2024;369:591–603.

    Article  CAS  PubMed  Google Scholar 

  250. Gyuris A, et al. Physical and molecular landscapes of mouse glioma extracellular vesicles define heterogeneity. Cell Rep. 2019;27(13):3972–e39876.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  251. Lokumcu T, et al. Proteomic, metabolomic, and fatty acid profiling of small extracellular vesicles from glioblastoma Stem-Like cells and their role in tumor heterogeneity. ACS Nano. 2024;18(3):2500–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  252. Tian Y, et al. Glioma-derived endothelial cells promote glioma cells migration via extracellular vesicles-mediated transfer of MYO1C. Biochem Biophys Res Commun; 2020.

  253. Lan F, et al. Serum Exosomal miR-301a as a potential diagnostic and prognostic biomarker for human glioma. Cell Oncol (Dordr). 2018;41(1):25–33.

    Article  CAS  PubMed  Google Scholar 

  254. Chen X, et al. MiR-9 promotes tumorigenesis and angiogenesis and is activated by MYC and OCT4 in human glioma. J Exp Clin Cancer Res. 2019;38(1):99.

    Article  PubMed  PubMed Central  Google Scholar 

  255. Shao N, et al. miR-454-3p is an Exosomal biomarker and functions as a tumor suppressor in glioma. Mol Cancer Ther. 2019;18(2):459–69.

    Article  CAS  PubMed  Google Scholar 

  256. Bian EB, et al. Exosomal lncRNA–ATB activates astrocytes that promote glioma cell invasion. Int J Oncol. 2019;54(2):713–21.

    CAS  PubMed  Google Scholar 

  257. Chai Y, et al. Exosomal LncRNA ROR1-AS1 derived from tumor cells promotes glioma progression via regulating miR-4686. Int J Nanomed. 2020;15:8863–72.

    Article  CAS  Google Scholar 

  258. Zhang Z, et al. Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to Temozolomide in glioblastoma. J Exp Clin Cancer Res. 2019;38(1):166.

    Article  PubMed  PubMed Central  Google Scholar 

  259. Graner MW, Cumming RI, Bigner DD. The heat shock response and chaperones/heat shock proteins in brain tumors: surface expression, release, and possible immune consequences. J Neurosci. 2007;27(42):11214–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  260. Hu P, et al. Exosomal HMGB3 released by glioma cells confers the activation of NLRP3 inflammasome and pyroptosis in tumor-associated macrophages. Tissue Cell. 2024;88:102406.

    Article  CAS  PubMed  Google Scholar 

  261. Pan Z, et al. EWSR1-induced circNEIL3 promotes glioma progression and exosome-mediated macrophage immunosuppressive polarization via stabilizing IGF2BP3. Mol Cancer. 2022;21(1):16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  262. Guo X, et al. Glioma exosomes mediate the expansion and function of myeloid-derived suppressor cells through microRNA-29a/Hbp1 and microRNA-92a/Prkar1a pathways. Int J Cancer. 2019;144(12):3111–26.

    Article  CAS  PubMed  Google Scholar 

  263. Huang K, et al. The role of PTRF/Cavin1 as a biomarker in both glioma and serum exosomes. Theranostics. 2018;8(6):1540–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  264. Mallawaaratchy DM, et al. Comprehensive proteome profiling of glioblastoma-derived extracellular vesicles identifies markers for more aggressive disease. J Neurooncol. 2017;131(2):233–44.

    Article  CAS  PubMed  Google Scholar 

  265. Ricklefs FL, et al. Imaging flow cytometry facilitates multiparametric characterization of extracellular vesicles in malignant brain tumours. J Extracell Vesicles. 2019;8(1):1588555.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  266. Indira Chandran V, et al. Ultrasensitive Immunoprofiling of plasma extracellular vesicles identifies Syndecan-1 as a potential tool for minimally invasive diagnosis of glioma. Clin Cancer Res. 2019;25(10):3115–27.

    Article  PubMed  Google Scholar 

  267. Lewis J, et al. A pilot Proof-Of-Principle analysis demonstrating dielectrophoresis (DEP) as a glioblastoma biomarker platform. Sci Rep. 2019;9(1):10279.

    Article  PubMed  PubMed Central  Google Scholar 

  268. Osti D, et al. Clinical significance of extracellular vesicles in plasma from glioblastoma patients. Clin Cancer Res. 2019;25(1):266–76.

    Article  CAS  PubMed  Google Scholar 

  269. Sabbagh Q, et al. The von Willebrand factor stamps plasmatic extracellular vesicles from glioblastoma patients. Sci Rep. 2021;11(1):22792.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  270. Ricklefs FL et al. FASN is a biomarker enriched in malignant Glioma-Derived extracellular vesicles. Int J Mol Sci, 2020. 21(6).

  271. Muller Bark J, et al. Proteome profiling of salivary small extracellular vesicles in glioblastoma patients. Cancer. 2023;129(18):2836–47.

    Article  PubMed  Google Scholar 

  272. Rana R, et al. Plasma-Derived extracellular vesicles reveal Galectin-3 binding protein as potential biomarker for early detection of glioma. Front Oncol. 2021;11:778754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  273. Chen WW, et al. BEAMing and droplet digital PCR analysis of mutant IDH1 mRNA in glioma patient serum and cerebrospinal fluid extracellular vesicles. Mol Ther Nucleic Acids. 2013;2(7):e109.

    Article  PubMed  PubMed Central  Google Scholar 

  274. Shao H, et al. Chip-based analysis of Exosomal mRNA mediating drug resistance in glioblastoma. Nat Commun. 2015;6:6999.

    Article  CAS  PubMed  Google Scholar 

  275. Manterola L, et al. A small noncoding RNA signature found in exosomes of GBM patient serum as a diagnostic tool. Neuro Oncol. 2014;16(4):520–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  276. Noerholm M, et al. RNA expression patterns in serum microvesicles from patients with glioblastoma multiforme and controls. BMC Cancer. 2012;12:22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  277. Muller L, et al. Exosomes isolated from plasma of glioma patients enrolled in a vaccination trial reflect antitumor immune activity and might predict survival. Oncoimmunology. 2015;4(6):e1008347.

    Article  PubMed  PubMed Central  Google Scholar 

  278. Tabibkhooei A, et al. Profiling of novel Circulating MicroRNAs as a non-invasive biomarker in diagnosis and follow-up of high and low-grade gliomas. Clin Neurol Neurosurg. 2020;190:105652.

    Article  PubMed  Google Scholar 

  279. García-Romero N, et al. DNA sequences within glioma-derived extracellular vesicles can cross the intact blood-brain barrier and be detected in peripheral blood of patients. Oncotarget. 2017;8(1):1416–28.

    Article  PubMed  Google Scholar 

  280. Manley GT, et al. Aquaporin-4 deletion in mice reduces brain edema after acute water intoxication and ischemic stroke. Nat Med. 2000;6(2):159–63.

    Article  CAS  PubMed  Google Scholar 

  281. Feng T, et al. Neuroprotective influence of miR-301a Inhibition in experimental cerebral ischemia/reperfusion rat models through targeting NDRG2. J Mol Neurosci. 2019;68(1):144–52.

    Article  CAS  PubMed  Google Scholar 

  282. Ou J et al. MiR-375 attenuates injury of cerebral ischemia/reperfusion via targetting Ctgf. Biosci Rep, 2017. 37(6).

  283. Wang HJ, et al. Catalpol improves impaired neurovascular unit in ischemic stroke rats via enhancing VEGF-PI3K/AKT and VEGF-MEK1/2/ERK1/2 signaling. Acta Pharmacol Sin. 2022;43(7):1670–85.

    Article  CAS  PubMed  Google Scholar 

  284. Zhu Y, et al. Angiopoietin-2 facilitates vascular endothelial growth factor-induced angiogenesis in the mature mouse brain. Stroke. 2005;36(7):1533–7.

    Article  CAS  PubMed  Google Scholar 

  285. Zacharek A, et al. Angiopoietin1/Tie2 and VEGF/Flk1 induced by MSC treatment amplifies angiogenesis and vascular stabilization after stroke. J Cereb Blood Flow Metab. 2007;27(10):1684–91.

    Article  CAS  PubMed  Google Scholar 

  286. Chen J, et al. Intravenous administration of human bone marrow stromal cells induces angiogenesis in the ischemic boundary zone after stroke in rats. Circ Res. 2003;92(6):692–9.

    Article  CAS  PubMed  Google Scholar 

  287. Qin LZ, et al. PTX3 expression in the plasma of elderly ACI patients and its relationship with severity and prognosis of the disease. Eur Rev Med Pharmacol Sci. 2016;20(19):4112–8.

    PubMed  Google Scholar 

  288. Yao H, et al. Hyperforin promotes Post-stroke neuroangiogenesis via astrocytic IL-6-Mediated negative immune regulation in the ischemic brain. Front Cell Neurosci. 2019;13:201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  289. Bang OY, et al. Caveolin-1, ring finger protein 213, and endothelial function in Moyamoya disease. Int J Stroke. 2016;11(9):999–1008.

    Article  PubMed  Google Scholar 

  290. Jiang H, et al. Differential expression of Circulating Exosomal MicroRNAs in refractory intracranial atherosclerosis associated with antiangiogenesis. Sci Rep. 2019;9(1):19429.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  291. Wen L, et al. The predictive role of early inflammation and oxidative stress and the dynamics of cytokines networks in post-stroke depression. J Affect Disord. 2024;347:469–76.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

No funding was received for this study.

Author information

Authors and Affiliations

Authors

Contributions

Zhongnan Hao wrote the main manuscript text and prepared Table 1, and 2; Wenxin Guan was responsible for revising the manuscript during the major revision; Wei Wei prepared Table 3, and 4; Meihua Li, Zhipeng Xiao, and Qinjian Sun prepared Figs. 1, 2 and 3; Zhongnan Hao, Yongli Pan and Wenqiang Xin concepted the manusctrip. All authors reviewed the manuscript.

Corresponding authors

Correspondence to Yongli Pan or Wenqiang Xin.

Ethics declarations

Ethical approval

Not applicable.

Competing interests

The authors declare 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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hao, Z., Guan, W., Wei, W. et al. Unlocking the therapeutic potential of tumor-derived EVs in ischemia-reperfusion: a breakthrough perspective from glioma and stroke. J Neuroinflammation 22, 84 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-025-03405-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12974-025-03405-7

Keywords