Next Article in Journal
Recent Advances in Co-Former Screening and Formation Prediction of Multicomponent Solid Forms of Low Molecular Weight Drugs
Next Article in Special Issue
Thermo-Responsive Hydrogels Encapsulating Targeted Core–Shell Nanoparticles as Injectable Drug Delivery Systems
Previous Article in Journal
Study of the Early Effects of Chitosan Nanoparticles with Glutathione in Rats with Osteoarthrosis
Previous Article in Special Issue
Design of Dual-Targeted pH-Sensitive Hybrid Polymer Micelles for Breast Cancer Treatment: Three Birds with One Stone
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Engineered Extracellular Vesicles for Drug Delivery in Therapy of Stroke

by
Waqas Ahmed
1,2,
Muhammed Shibil Kuniyan
2,
Aqil Mohammad Jawed
2 and
Lukui Chen
1,*
1
Department of Neurosurgery, Neuroscience Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510310, China
2
School of Medicine, Southeast University, Nanjing 210009, China
*
Author to whom correspondence should be addressed.
Pharmaceutics 2023, 15(9), 2173; https://doi.org/10.3390/pharmaceutics15092173
Submission received: 18 July 2023 / Revised: 10 August 2023 / Accepted: 17 August 2023 / Published: 22 August 2023

Abstract

:
Extracellular vesicles (EVs) are promising therapeutic modalities for treating neurological conditions. EVs facilitate intercellular communication among brain cells under normal and abnormal physiological conditions. The potential capability of EVs to pass through the blood–brain barrier (BBB) makes them highly promising as nanocarrier contenders for managing stroke. EVs possess several potential advantages compared to existing drug-delivery vehicles. These advantages include their capacity to surpass natural barriers, target specific cells, and stability within the circulatory system. This review explores the trafficking and cellular uptake of EVs and evaluates recent findings in the field of EVs research. Additionally, an overview is provided of the techniques researchers utilize to bioengineer EVs for stroke therapy, new results on EV–BBB interactions, and the limitations and prospects of clinically using EVs for brain therapies. The primary objective of this study is to provide a comprehensive analysis of the advantages and challenges related to engineered EVs drug delivery, specifically focusing on their application in the treatment of stroke.

1. Introduction

EVs are nanoscale heterogeneous structures released constitutively into the extracellular space by nearly all prokaryotic and eukaryotic cell types [1,2,3,4]. EVs are lipidic vesicles that develop in cells naturally. They facilitate cell-to-cell interaction by transferring various bioactive molecules from the mother cell, such as microRNAs, messenger RNAs, long noncoding RNAs (LncRNAs), circular RNAs (circRNAs) proteins, and lipids [5] (Figure 1). They are divided into three distinct categories: (i) Exosomes, (ii) Microvesicles, and (iii) Apoptotic bodies (Figure 2). Exosomes are a minor type of EVs, and prior electron microscopy studies have shown that they have a cup-like structure with a diameter ranging from 40 to 100 nm. During the development of multivesicular endosomes, an inward budding of the endosomal membrane produces exosomes [6]. Microvesicles, the second most significant type of vesicle, are formed when the plasma membrane buds and splits apart, with diameters between 100 and 1000 nm [7]. Apoptotic bodies represent the most abundant type of vesicles, exhibiting a size range of 1–5 μm and a diverse range of morphological characteristics. They are generated during the process of apoptosis. As a result, they possess a wide variety of constituents inherited from their progenitor cells, such as organelles and fragments of DNA [8].
Stroke is a prevalent neurological condition that can lead to permanent disability [9,10]. Annually, approximately 6.7 million individuals globally experience a stroke, with ischemic stroke comprising 87% of all mortalities [11]. The pathophysiological responses that occur after a stroke are complicated, and at present, tissue plasminogen activator remains the most efficacious treatment option for stroke [12,13], effective only when taken within 4–6 h of the onset of symptoms [14,15]. Despite this, due to its limited treatment window, it benefits less than 5% of patients [16]. Recent studies suggest that utilizing anti-inflammatory methods has considerable potential for expanding the time frame for treatment and decreasing severe brain damage following reperfusion [17,18]. Still, there is a constant demand for effective and cutting-edge medications that may prevent ischemia cascades and the diseases they cause.
EVs significantly function in the complex intercellular communication among neurons, glia, and vascular cells. They are essential in regulating homeostasis and influencing the development and prognosis of pathological conditions. EVs are involved in various physiological processes, including the maintenance and restoration of neurons [19], synaptic function [20], neurovascular stability [21], and the preservation of myelination [22]. In recent years, several studies have demonstrated the potential of EVs as nanotherapeutics for treating brain pathologies [23,24,25]. The scientific world has taken a keen interest in this topic, with numerous studies demonstrating the neuroprotective and regenerative effects of natural EVs from various resources [23,26,27].
The present review suggests that engineered EVs can enhance the curative effectiveness of EV-based treatments for stroke. We will explore the curative abilities of natural EVs for neurological applications, including crucial factors for their therapeutic success. We also discuss recent advances in regulating the BBB by EVs and their movement throughout the BBB. In our final part, we summarized the many scientific approaches discovered for modulating the content and surface of EVs, with a particular emphasis on the tactics employed for various therapeutic and targeting drugs.

2. Analysis Criteria

A digital database and search tool were utilized to thoroughly review scientific literature, including original articles regarding experimental and observational studies, case series, and reports, among other relevant sources. PubMed, Google Scholar, Scopus, Web of Science, bioRxiv, medRxiv, CNKI, and WanFang Data are central databanks utilized for medical studies. (The latter two databases are particularly significant within the Chinese mainland.) The study analyzed a total of 42 articles that were published or in preprint form between December 2017 and June 2023. A brief overview of the connection between EVs and stroke was found.

3. Isolation and Characterization of EVs

The primary challenge in isolating and characterizing EVs lies in their small size and heterogeneous nature. In 2018, the International Society for Extracellular Vesicles updated its “Minimum information for studies of extracellular vesicles” (MISEV) standards, which can be summed up as follows: (I) give a quantitative description of the EV source; (II) show the existence of the functioning bilayers of lipids and describe their purity; (III) use a mixture of EV analysis methods, ideally one optical and one biophysical/biochemical; and (IV) use a solid and accurate experimental design when figuring out how EVs work in the body [28,28].
The methodologies used for EV separation exhibit a wide range of diversity. Moreover, every single technique may possess unique parameters and configurations designed to enrich specific subtypes of EVs. The workflow of EV analysis is shown in (Figure 3), along with typical methods for EV size, concentration, and cargo measurement. The selection of the EV separation method, or a combination of methods, is significantly impacted by the research purpose, as well as considerations of time, costs, and applicability. This is particularly important when considering potential clinical applications [29].

4. EV Extraction from Biological Specimens

EVs have been extracted from a wide range of biological fluids, such as blood, urine, saliva, breast milk, cerebrospinal fluid, ascitic fluid, gastric juice, bile, sputum, bronchoalveolar lavage, semen, and tears [30,30,31,32,33,34]. They are actively released by cells throughout various tissues and organs, exhibiting their presence in physiological and pathological conditions [35,36]. The primary contributors to the population of blood microvesicles are platelets, which constitute a significant proportion of blood, with EVs ranging from 70% to 90% [35]. In addition to the mentioned hematopoietic cells, many other cell types, including reticulocytes, B lymphocytes, T cells, neutrophils, mast cells, dendritic cells, and macrophages, also play a significant role. Moreover, cells originating from different bodily tissues, such as epithelial cells, also contribute to this process [37]. The use of standardized pre-analytical procedures (Figure 3) is of the greatest significance to minimize the presence of mistakes in the analysis of EVs, mainly when these EVs are obtained from complex body fluids, such as blood. The composition, concentration, and characteristics of EVs produced from biofluids can be influenced by various factors, such as age, gender, ethnicity, body mass index, disease status, medication usage, overall lifestyle, and dietary lifestyles [38]. These variables must be considered and modified across all study participants (patients and any relevant controls).

5. Natural EVs for Treating Stroke

The significance of EV signaling within the brain’s framework was initially recorded in the early 1950s through electron microscopy studies [39,40,41,42]. Since then, researchers have discovered that EVs produced by nerve cells, like astrocytes and microglia, play an important role in immune signaling during synaptic plasticity [43,44,45]; in the specialized nature of neural cell interaction [46,47]; and in the diffusion of various neurological diseases, like neurodegenerative diseases and cerebral tumors [48]. Stroke [24], traumatic brain injury (TBI) [49], Alzheimer’s disease (AD) [50], autism [51], and schizophrenia [52] are just a few of the brain disorders where the therapeutic effects of EVs have been described since 2011. Over the past decade, a notable transition has been from cell-based treatments to EVs treatments. Several clinical investigations have exhibited the potential of EVs to protect and regenerate in various therapeutic applications in stroke (Table 1).

5.1. EV Origin

According to current research, the origin of EVs after systemic treatment affects their biodistribution [64]. This aspect has yet to be thoroughly investigated when designing EVs as therapies for stroke. A natural brain stereotype could be employed to enhance the therapeutic benefit of an EV. As far as current knowledge goes, research has yet to be conducted to directly contrast the brain tropism of extracellular vesicles derived from various origins. Research on native EVs for stroke has focused on their therapeutic potential rather than their specific ability to target the brain. According to experimental evidence, the mechanism by which EVs are derived from mesenchymal stem cells (MSCs) targeting damaged areas in the brain may be influenced by inflammatory processes [65]. Previous research has utilized EVs released by neural stem cells (NSCs) sourced from the subventricular area of mice [6,46,66] or from human NSCs [57] that were derived following the division of induced embryonic stem cells (Figure 4). Initial research utilizing mouse NSC-EVs indicated that these EVs are more likely to build in the liver and lung than the brain following intravenous or retro-orbital administration [55].
Substances on their surface mediate the natural targeting capability of EVs [67,68]. Findings from the report of metastatic progression have revealed that the brain tropism of EVs originating from breast tissue cells [68] is determined by expressing Integrin Beta3. Additional research on surface molecules that facilitate brain targeting may offer insights into selecting cell sources or developing engineering techniques to improve cerebral tropism.

5.2. Route of Administration

The route of administration is an essential factor to consider when investigating the biodistribution of a drug. This is also applicable to the investigation of EVs. (Figure 4). In a variety of animal experiments, EVs have been given to the animals intracerebrally [69,70,71,72,73], intravenously [74], intranasally [75], intra-arterially, intraperitoneally [76], and via retro-orbital [77] routes. Limited research has been conducted to compare the number of EVs produced in the nervous system through various delivery methods.
Administering EVs through the intra-arterial route is more productive for targeting the brain than the other routes. This is because the EVs are delivered near the brain, which reduces their elimination by other organs in the body [64]. EVs generated from human mesenchymal stem cells isolated from human bone marrow were administered intravenously to mice with acute brain lesions resembling ischemic stroke conditions [78]. Following treatment with EVs, a reduction in the presence of macrophages was observed in the affected area compared to the control group. In addition, there was a decline in the expression of pro-inflammatory cytokines and astrocyte activation.

5.3. EV Dose Comparisons

In addition to the delivery pathway and cellular origin, the number of EVs and the schedule of administration are critical factors that significantly impact the therapeutic effectiveness of the treatment (Figure 4). Various doses and administration regimens have been utilized to treat stroke using native EVs (Table 1). In stroke therapy, Aβ oligomers have been reduced in rats and a transgenic mouse model by administering native EVs at concentrations ranging from 10 to 100 μg and 30 to 30 μg, respectively [60,61,63,72,79]. Some studies administer EVs only once, while others administer them multiple times due to the quick elimination of EVs from the infarcted region within twenty-four hours of the initial intravenous injection [80] to achieve sustained brain accumulation. The administration time varied between 2 and 48 h post-ischemic stroke for single-dose therapies and 2 h weekly for multiple-dose regimens [23,60,81]. It has been suggested that reducing systemic inflammation in the blood after a stroke can be achieved by increasing the number of M2-type macrophages and Treg populations and decreasing the number of Th17 cells within 2 h of treatment. This creates a favorable environment for successful brain remodeling [57]. Importantly, from a therapeutic standpoint, higher concentrations of EVs are not always preferable. NPC-EVs or MSC-EVs at an average dose boosted neuronal densities in stroke mice but not at either low or high doses.

5.4. Mechanism of Action

The purpose of current study aims to examine the functional benefits of vesicles that are secreted from mesenchymal stem cells (MSCs) [82] and neural stem cells [57], which have been identified in rodents, rats, and pigs [60] models of cerebral ischemia that were caused by the blockage of the middle cerebral artery. It has been demonstrated that MSC-derived EVs have similar outcomes to those described with MSC transplantation in terms of decreasing infarct volume, enhancing functional recovery, increasing angiogenesis and neovascularization [83,84], decreasing astrocyte stimulation [60], and modulating peripheral immune system responses (Figure 5).
Most reported therapeutic effects of EVs in treating stroke are believed to be passive, mediated explicitly through extracranial organs. EVs facilitate a reduction in the overall inflammatory response following stroke, which might decrease leukocyte infiltration in the brain, ultimately reducing blood–brain permeation and neuronal inflammation [57,85]. The study reveals that NSC-EVs can induce the polarization of macrophages toward an M2 genotype with anti-inflammatory properties. Additionally, NSC-EVs were found to increase the population of regulatory T cells while reducing the number of proinflammatory T helper 17 cells [57].
The fact that EVs can cross the BBB and retain their functional cargo has significantly contributed to the development of biomarker research using EVs and their potential application as a vehicle for drug delivery. Alvarez-Erviti et al. showed that mice injected with EVs delivered siRNA to the brain. Dendritic cells were modified to express EVs membrane protein Lamp2b26. Genetically pairing Lamp2b with a CNS-specific rabies virus glycoprotein (RVG) peptide targeted EVs exclusively to the brain [50]. In another study, researchers utilized rats as subjects to investigate the presence of a fluorescently labeled protein specifically expressed in brain tissue. The study found this protein could be detected in microscopic EVs in the rats’ blood. This research study presents empirical support for intercellular communication facilitated by EVs originating from the CNS and disseminating throughout the peripheral tissues [86]. The studies provide evidence that supports the concept of EV crossing the BBB in a bidirectional manner. However, the precise mechanism by which this crossing occurs remains uncertain.
The therapeutic efficacy of EVs in stroke is primarily attributed to the molecular mechanisms involving microRNAs (miRNAs) present within these vesicles. Typically, in vitro experiments evaluate the direct impact of EVs on neuronal cells. They promote neurite outgrowth in nerve cells, and the inhibition of the growth factor for connective tissue in astrocytes was observed upon exposure to miR-133b-containing EVs released by mesenchymal stem cells (MSCs). This effect was attributed to the suppression of RhoA by the EVs [87,88]. Furthermore, it has been discovered that EVs containing miR-124, released by M2 microglia cells, can increase neuronal survival in vivo. The downregulation of ubiquitin-specific protease 14 is regulated to achieve the desired result. However, whether the effect is direct or systemic is still being determined since there is no apparent connection between transfected cells and the downregulation of ubiquitin-specific protease 14 (Figure 6).

6. Neuronal Regeneration via Sustained EV/NP Delivery

Over several decades, extensive research has been conducted on utilizing the body’s inherent regenerative capacity. Numerous studies have investigated the utilization of various therapeutic approaches to induce endogenous regeneration. These approaches include instructive biomaterial scaffolds, EVs, nanoparticles (NPs), small molecules, and other similar interventions [89]. Therapeutic EVs have been utilized for potential regenerative uses primarily due to their adaptability in modifying size and capacity to serve as carriers for transporting drugs, growth factors, tiny molecules, or genetic information [90,91].
Nerve injuries pose significant clinical challenges as a result of their inherent limitations in terms of regenerative capacity. Conventional treatments for nerve injuries have traditionally relied on autologous nerve grafts. However, these grafts are gradually being supplanted by artificial nerve guidance conduits (NGCs) due to limitations, such as the limited availability of nerve grafts, the need for multiple surgeries to isolate donor grafts, and the potential risk of developing neuromas. The primary objective of a recent research study on managing peripheral nerve injury was to explore the potential of targeted therapeutics. This was achieved by utilizing alginate hydrogels composed of laminin-coated poly(l-lactide-co-glycolide) (PLGA) conduits. These conduits were designed to contain a mixture of gold nanoparticles (NPs), brain-derived neurotropic growth factor (BDNF), and adipose-derived stem cells [92].
A separate investigation examined the administration of conductive poly(3,4-ethylenedioxythiophene) nanoparticles/EVs modified with cell adhesive tetrapeptide. These nanoparticles were delivered using a biocompatible chitin scaffold. The study’s results indicate that the conductive scaffold exhibited high porosity and compatibility with biological systems. Upon introduction into an in vivo model, a notable enhancement in nerve regeneration was observed, as evidenced by an increase in the regenerated myelin’s thickness and the muscle fibers’ area. An augmentation in the adhesive capacity of Schwann cells, along with enhanced angiogenesis, was also noted at the site of the injury (Figure 7). Hence, the utilization of this chitin scaffold with electrical activity has been proposed as a promising alternative for a nerve guidance conduit, as well as a suitable material for the administration of therapeutics to facilitate nerve regeneration [93].

7. Drug Loading Techniques

The effectiveness of EVs as a new class of nanocarriers arises from their unique qualities as information carriers, including their inherent homing ability, widespread distribution in biological fluids, biological compatibility, cell-specific targeting, non-immunogenicity, and easy penetration through physiological barriers. Pre-loading and post-loading are the two primary strategies for transferring cargo into EVs. Pre-loading refers to loading cargo into parent cells before the isolation of EVs, which releases o EVs that are loaded with cargo. Post-loading refers to loading cargo directly into EVs using either passive or active means after EVs have been isolated (Figure 8). Table 2 provides a comprehensive overview of various cargo loading tactics and procedures employed for extracellular vesicles, along with their advantages and disadvantages.

8. Engineered EVs for Stroke Treatment

Despite advancements in the preclinical application of endogenous EVs as therapeutic agents for stroke over the past decade, additional enhancements are required to optimize their therapeutic efficacy and expedite their clinical implementation. The first involves the biological effects of EVs. The EVs of native origin exhibit heterogeneity, even if obtained from an identical cell resource. Therefore, concentrating the EV material into a singular therapeutic entity with the most potent neuronal activity can enhance the efficacy of their treatment. The second is related to the focus on EV targeting. After being administered systemically, very few EVs (usually less than 5%) end up in the brain [109,110,111]. To have the most significant local or direct effect on the brain, we need to make strides in the surface engineering of EVs to improve their range and target particular cell membrane receptors. On the other hand, assessing the in vivo targeting and therapeutic mechanisms of EVs demands the creation of analytical and imaging platforms that are highly sensitive and have high resolution, respectively.
Consequently, engineering techniques to modify EV bioactivity [27,112,113,114,115,116,117,118,119,120,121], targeting [122,123,124,125,126,127,128,129,130], and tracking [123] have been created to tackle past difficulties. The earlier approaches can be implemented in EVs after isolation or within the cells responsible for EV production. This can be achieved through the use of gene editing [112], metabolic and residue-specific protein packaging [131], or the process of incubating cells with external compounds [132] or nanoparticles [133] (Figure 8 and Figure 9; Table 2 and Table 3). Numerous techniques are available for rapidly and efficiently engineering EVs with functional groups, genetic material, biologically active proteins, and peptides. However, the development of methods that do not adversely affect the function of EVs remains a challenging task. The present section investigates various methodologies, including genetics, exogenous delivery, and chemically inspired techniques, that can alter EVs’ outside appearance and composition to facilitate drug delivery to the brain.

8.1. Modulation of Content

Over the past five years, a remarkable increase in the number of described techniques for loading functional molecules into EVs has been seen [27,116,118,142,143]. The predominant approach employed in such methods involves genetic modification via plasmid transfection of the cells that secrete extracellular vesicles, thereby effectively regulating the composition of the EVs [27,114,116,119,144]. Plasmid transfection can be accomplished through electroporation or incubation with transfection reagents [117]. Therefore, EVs have modulated the expression of genes in human disease models, both in vitro and in vivo, by incorporating functional proteins, mRNAs, microRNAs (miRNAs), and other short noncoding RNAs [119,145]. The direct modulation technique has been implemented in manipulating isolated EVs to incorporate small drug molecules and for encapsulating proteins and small amounts of non-coding RNA within EVs (Figure 8) [106,146,147,148]. When implementing these strategies, it is essential to consider certain factors. The molecules’ size determines the material type that EVs can transport. Numerous molecules, such as miRNAs or small drugs, can be encapsulated within EVs. However, the capacity of EVs is restricted when it comes to larger molecules such as mRNA or proteins. Furthermore, a more significant amount of cargo sometimes equates with increased biological value. Additionally, genetically modified EVs’ effectiveness on the target cell is influenced by internalization efficiency, intracellular trafficking, and channels altered by the EV-based molecules.
The fact that these studies were performed on animal models is the only thing that could be considered a limitation. On the other hand, there is no evidence of this therapeutic possibility in the human field due to the challenges involved in the preparation of engineered EVs and the challenges related to the methods of administration needed to achieve a significant enough result to be considered a valid therapeutic strategy. Although there are plenty of issues to be resolved, the development of standardized techniques and guidelines for EV engineering, isolation, and storage has brought EV-based therapies closer to being utilized for stroke and other neurological illnesses (Table 4).

8.1.1. Nanoparticle

EVs are being utilized to enclose tiny therapeutic particles, typically comprising a medicinal substance, with sizes ranging from 10 nm [123] to 150 nm [152]. This approach aims to enhance the transportation of such nanoparticles across the blood–brain barrier (BBB). The loading of EVs can be achieved through two distinct processes: The first involves isolating the EVs and subsequently subjecting them to an electroporation procedure with nanoparticles [123]. The second process entails transfecting EV-secreting cells with nanoparticles and loading the EVs [68]. Recent research has indicated that EVs exhibit comparable therapeutic efficacy to stem cells derived from ischemic stroke treatment [56,81,153,154]. One central area for improvement in utilizing exosomes is the inability to target the ischemic lesion within the brain specifically. It has been found that using magnetic nanovesicles (MNVs) made from mesenchymal stem cells (MSCs) and filled with iron oxide nanoparticles makes it easier to target the area of the brain that is ischemic. This is achieved by applying an external magnetic field, which facilitates magnetic navigation. Using magnetic navigation resulted in a 5.1-fold increase in the EVs’ capacity to target the ischemic lesion specifically. The MNVs were found to have a significant impact on reducing the infarct volume and enhancing motor function. Additionally, they were observed to stimulate a defensive response, promote vascular development, and prevent cell death in cerebral ischemia [88,155,156,157].
An animal stroke model utilized gold nanoparticles coated with glucose to facilitate non-invasive neurological imaging and EV tracking. This approach aided in identifying the most effective administration route and size parameter [158,159]. The diagnosis and treatment of numerous neurological disorders are impeded due to the limited ability of diagnostic agents to penetrate the blood–brain barrier (BBB) [160]. Following the idea of the Nature Biotechnology Group, which involved the production of EVs as vehicles for genetic treatment with selective targets in the brain [50], gold-coated vesicles targeted brain cells by enhancing nanoparticle permeability to cross the BBB [161].

8.1.2. Proteins

Determining the appropriate approach for encapsulating therapeutic proteins within EVs is based on their intended utilization. The method of transfecting cells that secrete extracellular vesicles (EVs) with plasmids is utilized to produce enzyme-loaded EVs, including catalase [106], Cre-recombinase [117], and the lysosomal enzyme tripeptidylpeptidase-1 (TPP-1) [76]. Following a stroke, there is a change in the proportions of pro-inflammatory and anti-inflammatory proteins, impacting the size of the infarct and the individual’s functional outcome [162,163]. EVs containing IFN-Ɣ have been utilized in treating neurodegenerative conditions such as multiple sclerosis. The study demonstrated that IFN-Ɣ-stimulated extracellular vesicles (EVs) decreased demyelination and neurological inflammation in a murine model [158,164]. They investigated the impact of TNFα and interleukin-1β cytokines on the molecular composition and release of astrocyte-derived extracellular vesicles. The findings indicate that vesicles derived from astrocytes treated with TNFα and IL-1β contained a high concentration of miR-125a-5p and miR-16-5p, which are known to target proteins that regulate neurotrophin signaling.
Furthermore, it was noted that EVs injected with cytokines reduced the neuronal expression of NTKR3 and Bcl2 [165]. The inflammatory cytokine IL-1β, which affects the brain’s inflammatory response to injury, was administered into the brain. An IL-1β injection into the striatum led to more Ly6b+ leukocytes going to the lesion site and more circulating vesicles in the plasma of mice compared to controls. IL-1β also caused astrocytes to release EVs, which quickly passed the BBB [166].
An additional study examined the pro-angiogenic effects of microglial BV2 cells that were polarized by IL-4 and lipopolysaccharide. The cells polarized by IL-4 demonstrated an increase in endothelial cell tube formation through the secretion of EVs. Furthermore, the miRNA-26a profile was observed to be higher in comparison to the LPS-polarized group [167]. Researchers found that preconditioning neural stem cells with IL-6, a proinflammatory cytokine that promotes prosurvival signaling, reduced ischemic injury in a mouse stroke model [167,168].

8.1.3. Small Non-Coding RNAs

The study of EVs in the context of nervous system disorders is a relatively developing area of research. The blood–brain barrier (BBB) is formed by the tightly joined endothelial cells of brain capillaries, which effectively limit the passage of small molecules that are lipid-insoluble (90–98%) into the brain [132,169,170]. Two distinct approaches can be employed for the loading of non-coding RNAs in EVs: (I) chemical (e.g., chemically induced transfection) [106,171] or physical (e.g., electroporation) [50,132] strategies following EV separation; and (II) the introduction of plasmid-encoded non-coding RNAs or non-coding RNAs directly into EV-secreting cells through transfection [113,119,149].
The researcher Yang et al. employed EVs to transport circular RNA to the ischemic area of stroke. The study’s authors used a targeted approach to deliver Circ-SCMHI RNA to neuronal cells. This was achieved by expressing RVG peptides on exosome membranes, which served as a cargo-delivering vehicle system for the Circ-SCMHI RNA of interest. The RVG-circSCMH1-EVs have been found to enhance neuronal plasticity through their binding to MeCP2 and subsequent upregulation of downstream gene expression (Mobp, Igfbp3, Fxyd1, and Prodh). This mechanism is believed to contribute to the maintenance of proper brain function. The administration of RVG-CircSCMH1-EVs via intravenous injection enhanced motor recovery, digit movement, and functional recovery in rodent and non-human primate experimental models. The authors proposed that RVG-CircSCMH1-EVs may possess a broader therapeutic window compared to existing treatments, as it could be administered up to 24 h following the onset of stroke [172].
MiR-124 is recognized for its propensity to promote neuronal development in the developing and mature brain. EVs containing miR-124 facilitate the acquisition of neuronal identity by cortical neural progenitors and confer post-ischemic recovery by promoting reliable cortical neurogenesis [132]. The BACE1 gene was effectively knocked down through siRNA delivery via EVs. This resulted in a significant decrease in β-amyloid levels in the brains of mice with wild-type mutations [50]. MiRNA-210 exhibits promising prospects for enhancing angiogenesis in the context of brain area restoration after ischemic events. The enhancement of miRNA-210 expression resulted in a more excellent restoration of function after stroke. The delivery of miRNA-210 to the ischemic area was achieved by linking EVs with the c(RGDyK) peptide and adding miRNA-210.
This resulted in an upregulation of miR-210 at the ischemic site and increased the expression of integrinβ3, vascular endothelial growth factor, and CD34, significantly improving animal survival rates [130].

8.1.4. Neurotrophic Factors (NTFs)

The neurotrophic factor is being researched as a protective factor in neurological diseases [173,174]. They control the development of neural stem cells, which are accountable for restoring neurological function and repairing vascular damage caused by stroke [175,176]. Neurotrophic factors (NTFs) are a promising therapeutic option for stroke repair. Their diverse neuroprotective functions following ischemic events have been acknowledged [177,178]. The clinical application of NTF is currently not feasible due to the absence of an effective method for delivering it systemically to the ischemic area. Brain-derived neurotrophic factor (BDNF) is extensively studied for its neuroprotective and anti-inflammatory effects. BDNF was packed within naive exosomes and administered intravenously to rats to combat brain inflammation [179]. MiR-206 knockdown EVs reduced early damage to the brain by increasing BDNF levels following the treatment [180]. This suggests that bioengineering vesicles with BDNF could help with brain diseases like stroke. More research is needed to understand how EVs and NTFs work in stroke treatment. It would be highly beneficial to prioritize exploring this area in the future.

9. Conclusions and Future Prospective

The domain of EVs is presently considered a rapidly advancing field in fundamental science and applied research [181]. EVs derived from various cellular origins have demonstrated therapeutic efficacy in stroke. Over the past decade, significant progress has been achieved in the field (i) by demonstrating the curative value of specific groups of EVs in preclinical studies for cognitive and behavioral brain diseases; (ii) by discovering the systemic [57] and local [61] actions that characterize EV brain regeneration processes; (iii) by showing the effects of EVs in neurogenesis [132], neural protection, angiogenesis, and cerebral remodeling; and (iv) by describing how EVs are transported through the BBB [132,182]. Based on empirical evidence, EVs are swiftly eliminated from the brain, leading to a significant absence of EVs within a short time. So, experts are working on bioengineering EVs to improve their half-life in circulation, make them more available at the disease site, make it easier to deliver them to specific cells, and use them to provide therapeutic molecules or regenerative medicine.
According to a study [57], treating ischemic stroke with innate EVs resulted in particular outcomes, primarily attributed to the alteration of the immune response rather than a localized effect in the brain. However, high growth in the lesion area could produce a greater neuroprotective and pro-angiogenic impact. Numerous investigations have been conducted, primarily in vitro, wherein the culture environment may influence the EV’s biochemical and biophysical characteristics. Consequently, further research is needed to comprehend the engineered EVs physiological impacts on human well-being comprehensively.
In short, EVs possess significant potential for therapeutic applications. Clinical trials have exclusively involved endogenous EVs, while engineered EVs designed to target the brain have yet to undergo clinical testing. This is primarily due to challenges associated with the large-scale production of EVs and the production of EVs with similar characteristics. Collectively, EV-derived therapeutics and diagnostics have exhibited encouraging outcomes in diverse phases of clinical trials. Nevertheless, most clinical trials on EVs are underway, and the available published data regarding their current progress or results could be much better.
Translating modified EVs into clinical settings presents significant obstacles, including restricted techniques for versatile isolation and purification, EV variability, poor storage conditions, potential immunogenic adverse effects, and batch-to-batch variations. Therefore, much work must be done to utilize EVs’ therapeutic effects properly.

Author Contributions

W.A. conceived, designed, and wrote the manuscript. M.S.K. and A.M.J. provided critical revision and helped in the analysis of the manuscript. L.C. supervised and contributed to the concepts, the perspectives, and the discussion of ideas, helped in the correction, and proofread the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was sponsored by the National Key R&D Program of China (2022YFA1104900 and 2022YFA1104904), the Natural Science Foundation of Guangdong Province (2021A1515010013), the Department of Education of Guangdong Province (2021ZDZX2011), the Traditional Chinese Medicine Bureau of Guangdong Province (20221275), the Guangzhou Municipal Science and Technology Project (202201011760), and the President Foundation of the Integrated Hospital of Traditional Chinese Medicine of Southern Medical University (1202101003).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Brown, L.; Wolf, J.M.; Prados-Rosales, R.; Casadevall, A. Through the wall: Extracellular vesicles in Gram-positive bacteria, mycobacteria and fungi. Nat. Rev. Microbiol. 2015, 13, 620–630. [Google Scholar] [CrossRef]
  2. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
  3. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef]
  4. Schwechheimer, C.; Kuehn, M.J. Outer-membrane vesicles from Gram-negative bacteria: Biogenesis and functions. Nat. Rev. Microbiol. 2015, 13, 605–619. [Google Scholar] [CrossRef]
  5. Kalluri, R.; LeBleu, V.S. The biology, function, and biomedical applications of exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef] [PubMed]
  6. Van Niel, G.; d’Angelo, G.; Raposo, G. Shedding light on the cell biology of extracellular vesicles. Nat. Rev. Mol. Cell Biol. 2018, 19, 213–228. [Google Scholar] [CrossRef]
  7. Gharbi, T.; Zhang, Z.; Yang, G.-Y. The function of astrocyte mediated extracellular vesicles in central nervous system diseases. Front. Cell Dev. Biol. 2020, 8, 568889. [Google Scholar] [CrossRef]
  8. Lu, M.; DiBernardo, E.; Parks, E.; Fox, H.; Zheng, S.-Y.; Wayne, E. The role of extracellular vesicles in the pathogenesis and treatment of autoimmune disorders. Front. Immunol. 2021, 12, 566299. [Google Scholar] [CrossRef]
  9. Murphy, T.H.; Corbett, D. Plasticity during stroke recovery: From synapse to behaviour. Nat. Rev. Neurosci. 2009, 10, 861–872. [Google Scholar] [CrossRef]
  10. Dimyan, M.A.; Cohen, L.G. Neuroplasticity in the context of motor rehabilitation after stroke. Nat. Rev. Neurol. 2011, 7, 76–85. [Google Scholar] [CrossRef]
  11. Cunningham, C.J.; Wong, R.; Barrington, J.; Tamburrano, S.; Pinteaux, E.; Allan, S.M. Systemic conditioned medium treatment from interleukin-1 primed mesenchymal stem cells promotes recovery after stroke. Stem Cell Res. Ther. 2020, 11, 1–12. [Google Scholar] [CrossRef] [PubMed]
  12. Otero-Ortega, L.; Laso-García, F.; Gómez-de Frutos, M.; Fuentes, B.; Diekhorst, L.; Díez-Tejedor, E.; Gutiérrez-Fernández, M. Role of exosomes as a treatment and potential biomarker for stroke. Transl. Stroke Res. 2019, 10, 241–249. [Google Scholar] [CrossRef] [PubMed]
  13. Pan, J.; Qu, M.; Li, Y.; Wang, L.; Zhang, L.; Wang, Y.; Tang, Y.; Tian, H.-L.; Zhang, Z.; Yang, G.-Y. MicroRNA-126-3p/-5p overexpression attenuates blood-brain barrier disruption in a mouse model of middle cerebral artery occlusion. Stroke 2020, 51, 619–627. [Google Scholar] [CrossRef] [PubMed]
  14. Malone, K.; Amu, S.; Moore, A.C.; Waeber, C. The immune system and stroke: From current targets to future therapy. Immunol. Cell Biol. 2019, 97, 5–16. [Google Scholar] [CrossRef]
  15. Wang, J.; Liu, H.; Chen, S.; Zhang, W.; Chen, Y.; Yang, Y. Moderate exercise has beneficial effects on mouse ischemic stroke by enhancing the functions of circulating endothelial progenitor cell-derived exosomes. Exp. Neurol. 2020, 330, 113325. [Google Scholar] [CrossRef]
  16. Wang, F.; Tang, H.; Zhu, J.; Zhang, J.H. Transplanting mesenchymal stem cells for treatment of ischemic stroke. Cell Transplant. 2018, 27, 1825–1834. [Google Scholar] [CrossRef]
  17. Iadecola, C.; Anrather, J. The immunology of stroke: From mechanisms to translation. Nat. Med. 2011, 17, 796–808. [Google Scholar] [CrossRef]
  18. Kelly, P.J.; Murphy, S.; Coveney, S.; Purroy, F.; Lemmens, R.; Tsivgoulis, G.; Price, C. Anti-inflammatory approaches to ischaemic stroke prevention. J. Neurol. Neurosurg. Psychiatry 2018, 89, 211–218. [Google Scholar] [CrossRef]
  19. Pascua-Maestro, R.; González, E.; Lillo, C.; Ganfornina, M.D.; Falcón-Pérez, J.M.; Sanchez, D. Extracellular vesicles secreted by astroglial cells transport apolipoprotein D to neurons and mediate neuronal survival upon oxidative stress. Front. Cell. Neurosci. 2019, 12, 526. [Google Scholar] [CrossRef]
  20. Antonucci, F.; Turola, E.; Riganti, L.; Caleo, M.; Gabrielli, M.; Perrotta, C.; Novellino, L.; Clementi, E.; Giussani, P.; Viani, P. Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J. 2012, 31, 1231–1240. [Google Scholar] [CrossRef]
  21. Xu, B.; Zhang, Y.; Du, X.-F.; Li, J.; Zi, H.-X.; Bu, J.-W.; Yan, Y.; Han, H.; Du, J.-L. Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Res. 2017, 27, 882–897. [Google Scholar] [CrossRef] [PubMed]
  22. Prada, I.; Gabrielli, M.; Turola, E.; Iorio, A.; D’Arrigo, G.; Parolisi, R.; De Luca, M.; Pacifici, M.; Bastoni, M.; Lombardi, M. Glia-to-neuron transfer of miRNAs via extracellular vesicles: A new mechanism underlying inflammation-induced synaptic alterations. Acta Neuropathol. 2018, 135, 529–550. [Google Scholar] [CrossRef] [PubMed]
  23. Webb, R.L.; Kaiser, E.E.; Jurgielewicz, B.J.; Spellicy, S.; Scoville, S.L.; Thompson, T.A.; Swetenburg, R.L.; Hess, D.C.; West, F.D.; Stice, S.L. Human neural stem cell extracellular vesicles improve recovery in a porcine model of ischemic stroke. Stroke 2018, 49, 1248–1256. [Google Scholar] [CrossRef] [PubMed]
  24. Xin, H.; Li, Y.; Cui, Y.; Yang, J.J.; Zhang, Z.G.; Chopp, M. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J. Cereb. Blood Flow Metab. 2013, 33, 1711–1715. [Google Scholar] [CrossRef]
  25. Feng, J.; Waqas, A.; Zhu, Z.; Chen, L. Exosomes: Applications in respiratory infectious diseases and prospects for coronavirus disease 2019 (COVID-19). J. Biomed. Nanotechnol. 2020, 16, 399–418. [Google Scholar] [CrossRef]
  26. Upadhya, D.; Shetty, A.K. Extracellular vesicles as therapeutics for brain injury and disease. Curr. Pharm. Des. 2019, 25, 3500–3505. [Google Scholar] [CrossRef] [PubMed]
  27. Sun, X.; Jung, J.-H.; Arvola, O.; Santoso, M.R.; Giffard, R.G.; Yang, P.C.; Stary, C.M. Stem cell-derived exosomes protect astrocyte cultures from in vitro ischemia and decrease injury as post-stroke intravenous therapy. Front. Cell. Neurosci. 2019, 13, 394. [Google Scholar] [CrossRef] [PubMed]
  28. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; 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, 1535750. [Google Scholar] [CrossRef]
  29. Sáenz-Cuesta, M.; Arbelaiz, A.; Oregi, A.; Irizar, H.; Osorio-Querejeta, I.; Muñoz-Culla, M.; Banales, J.M.; Falcón-Pérez, J.M.; Olascoaga, J.; Otaegui, D. Methods for extracellular vesicles isolation in a hospital setting. Front. Immunol. 2015, 6, 50. [Google Scholar] [CrossRef]
  30. Caby, M.P.; Lankar, D.; Vincendeau-Scherrer, C.; Raposo, G.; Bonnerot, C. Exosomal-like vesicles are present in human blood plasma. Int. Immunol. 2005, 17, 879–887. [Google Scholar] [CrossRef]
  31. Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  32. Akers, J.C.; Ramakrishnan, V.; Kim, R.; Phillips, S.; Kaimal, V.; Mao, Y.; Hua, W.; Yang, I.; Fu, C.C.; Nolan, J.; et al. miRNA contents of cerebrospinal fluid extracellular vesicles in glioblastoma patients. J. Neuro-Oncol. 2015, 123, 205–216. [Google Scholar] [CrossRef] [PubMed]
  33. Choi, H.I.; Choi, J.P.; Seo, J.; Kim, B.J.; Rho, M.; Han, J.K.; Kim, J.G. Helicobacter pylori-derived extracellular vesicles increased in the gastric juices of gastric adenocarcinoma patients and induced inflammation mainly via specific targeting of gastric epithelial cells. Exp. Mol. Med. 2017, 49, e330. [Google Scholar] [CrossRef]
  34. Wahlund, C.J.E.; Eklund, A.; Grunewald, J.; Gabrielsson, S. Pulmonary Extracellular Vesicles as Mediators of Local and Systemic Inflammation. Front. Cell Dev. Biol. 2017, 5, 39. [Google Scholar] [CrossRef] [PubMed]
  35. Żmigrodzka, M.; Guzera, M.; Miśkiewicz, A.; Jagielski, D.; Winnicka, A. The biology of extracellular vesicles with focus on platelet microparticles and their role in cancer development and progression. Tumor Biol. 2016, 37, 14391–14401. [Google Scholar] [CrossRef]
  36. Ciardiello, C.; Cavallini, L.; Spinelli, C.; Yang, J.; Reis-Sobreiro, M.; De Candia, P.; Minciacchi, V.R.; Di Vizio, D. Focus on extracellular vesicles: New frontiers of cell-to-cell communication in cancer. Int. J. Mol. Sci. 2016, 17, 175. [Google Scholar] [CrossRef]
  37. Frydrychowicz, M.; Kolecka-Bednarczyk, A.; Madejczyk, M.; Yasar, S.; Dworacki, G. Exosomes–structure, biogenesis and biological role in non-small-cell lung cancer. Scand. J. Immunol. 2015, 81, 2–10. [Google Scholar] [CrossRef]
  38. Barteneva, N.S.; Fasler-Kan, E.; Bernimoulin, M.; Stern, J.N.; Ponomarev, E.D.; Duckett, L.; Vorobjev, I.A. Circulating microparticles: Square the circle. BMC Cell Biol. 2013, 14, 15. [Google Scholar] [CrossRef]
  39. Maxwell, D.S.; Pease, D.C. The electron microscopy of the choroid plexus. J. Biophys. Biochem. Cytol. 1956, 2, 467. [Google Scholar] [CrossRef]
  40. Jorge, L.R. Morphological evidence for possible functional role of supra-ependymal nerves on ependyma. Brain Res. 1977, 125, 362–368. [Google Scholar] [CrossRef]
  41. Cupédo, R. The surface ultrastructure of the habenular complex of the rat. Anat. Embryol. 1977, 152, 43–64. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, L.; Zhang, G.; Feng, S.; Xue, M.; Cai, J.; Chen, L.; Deng, Y.; Wang, Y.; Chinese Association of Neurorestoratology (Preparatory); China Committee of International Association of Neurorestoratology. Preparation and quality control standard of clinical-grade neural progenitor/precursor cells-derived exosomes (2022 China version). J. Neurorestoratology 2022, 10, 100001. [Google Scholar] [CrossRef]
  43. Bianco, F.; Perrotta, C.; Novellino, L.; Francolini, M.; Riganti, L.; Menna, E.; Saglietti, L.; Schuchman, E.H.; Furlan, R.; Clementi, E. Acid sphingomyelinase activity triggers microparticle release from glial cells. EMBO J. 2009, 28, 1043–1054. [Google Scholar] [CrossRef] [PubMed]
  44. Potolicchio, I.; Carven, G.J.; Xu, X.; Stipp, C.; Riese, R.J.; Stern, L.J.; Santambrogio, L. Proteomic analysis of microglia-derived exosomes: Metabolic role of the aminopeptidase CD13 in neuropeptide catabolism. J. Immunol. 2005, 175, 2237–2243. [Google Scholar] [CrossRef]
  45. Belykh, E.; Shaffer, K.V.; Lin, C.; Byvaltsev, V.A.; Preul, M.C.; Chen, L. Blood-brain barrier, blood-brain tumor barrier, and fluorescence-guided neurosurgical oncology: Delivering optical labels to brain tumors. Front. Oncol. 2020, 10, 739. [Google Scholar] [CrossRef]
  46. Chivet, M.; Javalet, C.; Laulagnier, K.; Blot, B.; Hemming, F.J.; Sadoul, R. Exosomes secreted by cortical neurons upon glutamatergic synapse activation specifically interact with neurons. J. Extracell. Vesicles 2014, 3, 24722. [Google Scholar] [CrossRef]
  47. Chen, W.; Li, Q.; Zhang, G.; Wang, H.; Zhu, Z.; Chen, L. LncRNA HOXA-AS3 promotes the malignancy of glioblastoma through regulating miR-455-5p/USP3 axis. J. Cell. Mol. Med. 2020, 24, 11755–11767. [Google Scholar] [CrossRef]
  48. Budnik, V.; Ruiz-Cañada, C.; Wendler, F. Extracellular vesicles round off communication in the nervous system. Nat. Rev. Neurosci. 2016, 17, 160–172. [Google Scholar] [CrossRef]
  49. Zhang, Y.; Chopp, M.; Meng, Y.; Katakowski, M.; Xin, H.; Mahmood, A.; Xiong, Y. Effect of exosomes derived from multipluripotent mesenchymal stromal cells on functional recovery and neurovascular plasticity in rats after traumatic brain injury. J. Neurosurg. 2015, 122, 856–867. [Google Scholar] [CrossRef]
  50. Alvarez-Erviti, L.; Seow, Y.; Yin, H.; Betts, C.; Lakhal, S.; Wood, M.J. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 2011, 29, 341–345. [Google Scholar] [CrossRef]
  51. Geffen, Y.; Perets, N.; Horev, R.; Yudin, D.; Oron, O.; Elliott, E.; Marom, E.; Danon, U.; Offen, D. Exosomes derived from adipose mesenchymal stem cells: A potential non-invasive intranasal treatment for autism. Cytotherapy 2020, 22, S49. [Google Scholar] [CrossRef]
  52. Tsivion-Visbord, H.; Perets, N.; Sofer, T.; Bikovski, L.; Goldshmit, Y.; Ruban, A.; Offen, D. Mesenchymal stem cells derived extracellular vesicles improve behavioral and biochemical deficits in a phencyclidine model of schizophrenia. Transl. Psychiatry 2020, 10, 305. [Google Scholar] [CrossRef] [PubMed]
  53. Doeppner, T.R.; Herz, J.; Görgens, A.; Schlechter, J.; Ludwig, A.-K.; Radtke, S.; de Miroschedji, K.; Horn, P.A.; Giebel, B.; Hermann, D.M. Extracellular Vesicles Improve Post-Stroke Neuroregeneration and Prevent Postischemic Immunosuppression. Stem Cells Transl. Med. 2015, 4, 1131–1143. [Google Scholar] [CrossRef] [PubMed]
  54. Song, Y.; Li, Z.; He, T.; Qu, M.; Jiang, L.; Li, W.; Shi, X.; Pan, J.; Zhang, L.; Wang, Y.; et al. M2 microglia-derived exosomes protect the mouse brain from ischemia-reperfusion injury via exosomal miR-124. Theranostics 2019, 9, 2910–2923. [Google Scholar] [CrossRef]
  55. Zheng, X.; Zhang, L.; Kuang, Y.; Venkataramani, V.; Jin, F.; Hein, K.; Zafeiriou, M.P.; Lenz, C.; Moebius, W.; Kilic, E. Extracellular Vesicles Derived from Neural Progenitor Cells—A Preclinical Evaluation for Stroke Treatment in Mice. Transl. Stroke Res. 2021, 12, 185–203. [Google Scholar] [CrossRef] [PubMed]
  56. Long, J.; Gu, C.; Zhang, Q.; Liu, J.; Huang, J.; Li, Y.; Zhang, Y.; Li, R.; Ahmed, W.; Zhang, J. Extracellular vesicles from medicated plasma of Buyang Huanwu decoction-preconditioned neural stem cells accelerate neurological recovery following ischemic stroke. Front. Cell Dev. Biol. 2023, 11, 1096329. [Google Scholar] [CrossRef] [PubMed]
  57. Webb, R.L.; Kaiser, E.E.; Scoville, S.L.; Thompson, T.A.; Fatima, S.; Pandya, C.; Sriram, K.; Swetenburg, R.L.; Vaibhav, K.; Arbab, A.S. Human neural stem cell extracellular vesicles improve tissue and functional recovery in the murine thromboembolic stroke model. Transl. Stroke Res. 2018, 9, 530–539. [Google Scholar] [CrossRef]
  58. Chen, L. Exosomes Derived from Human Neural Stem Cell Improve Recovery in a Cynomolgus Monkey Model of Ischemic Stroke. J. Neurosurg. 2020, 132, 12. [Google Scholar]
  59. Chen, K.-H.; Chen, C.-H.; Wallace, C.G.; Yuen, C.-M.; Kao, G.-S.; Chen, Y.-L.; Shao, P.-L.; Chen, Y.-L.; Chai, H.-T.; Lin, K.-C. Intravenous administration of xenogenic adipose-derived mesenchymal stem cells (ADMSC) and ADMSC-derived exosomes markedly reduced brain infarct volume and preserved neurological function in rat after acute ischemic stroke. Oncotarget 2016, 7, 74537. [Google Scholar] [CrossRef]
  60. Lee, J.Y.; Kim, E.; Choi, S.-M.; Kim, D.-W.; Kim, K.P.; Lee, I.; Kim, H.-S. Microvesicles from brain-extract—Treated mesenchymal stem cells improve neurological functions in a rat model of ischemic stroke. Sci. Rep. 2016, 6, 1–14. [Google Scholar] [CrossRef]
  61. Mahdavipour, M.; Hassanzadeh, G.; Seifali, E.; Mortezaee, K.; Aligholi, H.; Shekari, F.; Sarkoohi, P.; Zeraatpisheh, Z.; Nazari, A.; Movassaghi, S. Effects of neural stem cell-derived extracellular vesicles on neuronal protection and functional recovery in the rat model of middle cerebral artery occlusion. Cell Biochem. Funct. 2020, 38, 373–383. [Google Scholar] [CrossRef] [PubMed]
  62. Lin, C.; Huang, S.; Zhang, J.; Yuan, H.; Yao, T.; Chen, L. Dl-3-N-butylphthalide attenuates hypoxic injury of neural stem cells by increasing hypoxia-inducible factor-1alpha. J. Stroke Cerebrovasc. Dis. 2022, 31, 106221. [Google Scholar] [CrossRef] [PubMed]
  63. Safakheil, M.; Safakheil, H. The effect of exosomes derived from bone marrow stem cells in combination with rosuvastatin on functional recovery and neuroprotection in rats after ischemic stroke. J. Mol. Neurosci. 2020, 70, 724–737. [Google Scholar] [CrossRef] [PubMed]
  64. Wiklander, O.P.; Nordin, J.Z.; O’Loughlin, A.; Gustafsson, Y.; Corso, G.; Mäger, I.; Vader, P.; Lee, Y.; Sork, H.; Seow, Y. Extracellular vesicle in vivo biodistribution is determined by cell source, route of administration and targeting. J. Extracell. Vesicles 2015, 4, 26316. [Google Scholar] [CrossRef]
  65. Perets, N.; Betzer, O.; Shapira, R.; Brenstein, S.; Angel, A.; Sadan, T.; Ashery, U.; Popovtzer, R.; Offen, D. Golden exosomes selectively target brain pathologies in neurodegenerative and neurodevelopmental disorders. Nano Lett. 2019, 19, 3422–3431. [Google Scholar] [CrossRef]
  66. Chen, W.; Wang, H.; Feng, J.; Chen, L. Overexpression of circRNA circUCK2 attenuates cell apoptosis in cerebral ischemia-reperfusion injury via miR-125b-5p/GDF11 signaling. Mol. Ther.-Nucleic Acids 2020, 22, 673–683. [Google Scholar] [CrossRef]
  67. Cossetti, C.; Iraci, N.; Mercer, T.R.; Leonardi, T.; Alpi, E.; Drago, D.; Alfaro-Cervello, C.; Saini, H.K.; Davis, M.P.; Schaeffer, J. Extracellular vesicles from neural stem cells transfer IFN-γ via Ifngr1 to activate Stat1 signaling in target cells. Mol. Cell 2014, 56, 193–204. [Google Scholar] [CrossRef]
  68. Hoshino, A.; Costa-Silva, B.; Shen, T.-L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef]
  69. Zheng, T.; Pu, J.; Chen, Y.; Mao, Y.; Guo, Z.; Pan, H.; Zhang, L.; Zhang, H.; Sun, B.; Zhang, B. Plasma exosomes spread and cluster around β-amyloid plaques in an animal model of Alzheimer’s disease. Front. Aging Neurosci. 2017, 9, 12. [Google Scholar] [CrossRef]
  70. Orefice, N.S.; Souchet, B.; Braudeau, J.; Alves, S.; Piguet, F.; Collaud, F.; Ronzitti, G.; Tada, S.; Hantraye, P.; Mingozzi, F. Real-time monitoring of exosome enveloped-AAV spreading by endomicroscopy approach: A new tool for gene delivery in the brain. Mol. Ther.-Methods Clin. Dev. 2019, 14, 237–251. [Google Scholar] [CrossRef]
  71. Elia, C.A.; Tamborini, M.; Rasile, M.; Desiato, G.; Marchetti, S.; Swuec, P.; Mazzitelli, S.; Clemente, F.; Anselmo, A.; Matteoli, M. Intracerebral injection of extracellular vesicles from mesenchymal stem cells exerts reduced Aβ plaque burden in early stages of a preclinical model of Alzheimer’s disease. Cells 2019, 8, 1059. [Google Scholar] [CrossRef] [PubMed]
  72. An, K.; Klyubin, I.; Kim, Y.; Jung, J.H.; Mably, A.J.; O’Dowd, S.T.; Lynch, T.; Kanmert, D.; Lemere, C.A.; Finan, G.M. Exosomes neutralize synaptic-plasticity-disrupting activity of Aβ assemblies in vivo. Mol. Brain 2013, 6, 1–47. [Google Scholar] [CrossRef] [PubMed]
  73. Gu, C.; Feng, J.; Waqas, A.; Deng, Y.; Zhang, Y.; Chen, W.; Long, J.; Huang, S.; Chen, L. Technological advances of 3D scaffold-based stem cell/exosome therapy in tissues and organs. Front. Cell Dev. Biol. 2021, 9, 709204. [Google Scholar] [CrossRef] [PubMed]
  74. Zhang, Y.; Chopp, M.; Zhang, Z.G.; Katakowski, M.; Xin, H.; Qu, C.; Ali, M.; Mahmood, A.; Xiong, Y. Systemic administration of cell-free exosomes generated by human bone marrow derived mesenchymal stem cells cultured under 2D and 3D conditions improves functional recovery in rats after traumatic brain injury. Neurochem. Int. 2017, 111, 69–81. [Google Scholar] [CrossRef] [PubMed]
  75. Betzer, O.; Perets, N.; Angel, A.; Motiei, M.; Sadan, T.; Yadid, G.; Offen, D.; Popovtzer, R. In vivo neuroimaging of exosomes using gold nanoparticles. ACS Nano 2017, 11, 10883–10893. [Google Scholar] [CrossRef] [PubMed]
  76. Haney, M.J.; Klyachko, N.L.; Harrison, E.B.; Zhao, Y.; Kabanov, A.V.; Batrakova, E.V. TPP1 delivery to lysosomes with extracellular vesicles and their enhanced brain distribution in the animal model of batten disease. Adv. Healthc. Mater. 2019, 8, 1801271. [Google Scholar] [CrossRef]
  77. Ni, H.; Yang, S.; Siaw-Debrah, F.; Hu, J.; Wu, K.; He, Z.; Yang, J.; Pan, S.; Lin, X.; Ye, H. Exosomes derived from bone mesenchymal stem cells ameliorate early inflammatory responses following traumatic brain injury. Front. Neurosci. 2019, 13, 14. [Google Scholar] [CrossRef]
  78. Dabrowska, S.; Andrzejewska, A.; Strzemecki, D.; Muraca, M.; Janowski, M.; Lukomska, B. Human bone marrow mesenchymal stem cell-derived extracellular vesicles attenuate neuroinflammation evoked by focal brain injury in rats. J. Neuroinflammation 2019, 16, 1–15. [Google Scholar] [CrossRef]
  79. Huang, H.; Bach, J.R.; Sharma, H.S.; Saberi, H.; Jeon, S.R.; Guo, X.; Shetty, A.; Hawamdeh, Z.; Sharma, A.; von Wild, K. The 2022 yearbook of Neurorestoratology. J. Neurorestoratology 2023, 11, 100054. [Google Scholar] [CrossRef]
  80. Aboody, K.S.; Brown, A.; Rainov, N.G.; Bower, K.A.; Liu, S.; Yang, W.; Small, J.E.; Herrlinger, U.; Ourednik, V.; Black, P.M. Neural stem cells display extensive tropism for pathology in adult brain: Evidence from intracranial gliomas. Proc. Natl. Acad. Sci. USA 2000, 97, 12846–12851. [Google Scholar] [CrossRef]
  81. Chen, W.; Wang, H.; Zhu, Z.; Feng, J.; Chen, L. Exosome-shuttled circSHOC2 from IPASs regulates neuronal autophagy and ameliorates ischemic brain injury via the miR-7670-3p/SIRT1 axis. Mol. Ther.-Nucleic Acids 2020, 22, 657–672. [Google Scholar] [CrossRef]
  82. Otero-Ortega, L.; Laso-García, F.; Gómez-de Frutos, M.d.C.; Rodríguez-Frutos, B.; Pascual-Guerra, J.; Fuentes, B.; Díez-Tejedor, E.; Gutiérrez-Fernández, M. White matter repair after extracellular vesicles administration in an experimental animal model of subcortical stroke. Sci. Rep. 2017, 7, 44433. [Google Scholar] [CrossRef] [PubMed]
  83. Zhao, T.; Sun, F.; Liu, J.; Ding, T.; She, J.; Mao, F.; Xu, W.; Qian, H.; Yan, Y. Emerging role of mesenchymal stem cell-derived exosomes in regenerative medicine. Curr. Stem Cell Res. Ther. 2019, 14, 482–494. [Google Scholar] [CrossRef] [PubMed]
  84. Lin, C.-Q.; Chen, L.-K. Effect of differential hypoxia-related gene expression on glioblastoma. J. Int. Med. Res. 2021, 49, 03000605211013774. [Google Scholar] [CrossRef]
  85. Gu, C.; Liu, J.; Li, Y.; Zhang, Q.; Lin, C.; Huang, J.; Duan, W.; Deng, Y.; Ahmed, W.; Li, R. Comparison of ketamine/xylazine and isoflurane anesthesia on the establishment of mouse middle cerebral artery occlusion model. Exp. Anim. 2023, 72, 209–217. [Google Scholar] [CrossRef] [PubMed]
  86. Gómez-Molina, C.; Sandoval, M.; Henzi, R.; Ramírez, J.P.; Varas-Godoy, M.; Luarte, A.; Lafourcade, C.A.; Lopez-Verrilli, A.; Smalla, K.-H.; Kaehne, T. Small extracellular vesicles in rat serum contain astrocyte-derived protein biomarkers of repetitive stress. Int. J. Neuropsychopharmacol. 2019, 22, 232–246. [Google Scholar] [CrossRef]
  87. Xin, H.; Li, Y.; Buller, B.; Katakowski, M.; Zhang, Y.; Wang, X.; Shang, X.; Zhang, Z.G.; Chopp, M. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 2012, 30, 1556–1564. [Google Scholar] [CrossRef]
  88. Yu, Y.; Zheng, Y.; Dong, X.; Qiao, X.; Tao, Y. Efficacy and safety of tirofiban in patients with acute ischemic stroke without large-vessel occlusion and not receiving intravenous thrombolysis: A randomized controlled open-label trial. J. Neurorestoratology 2022, 10, 100026. [Google Scholar] [CrossRef]
  89. van Rijt, S.; Habibovic, P. Enhancing regenerative approaches with nanoparticles. J. R. Soc. Interface 2017, 14, 20170093. [Google Scholar] [CrossRef]
  90. Saleh, B.; Dhaliwal, H.K.; Portillo-Lara, R.; Shirzaei Sani, E.; Abdi, R.; Amiji, M.M.; Annabi, N. Local immunomodulation using an adhesive hydrogel loaded with miRNA-laden nanoparticles promotes wound healing. Small 2019, 15, 1902232. [Google Scholar] [CrossRef]
  91. Tan, H.-L.; Teow, S.-Y.; Pushpamalar, J. Application of metal nanoparticle–hydrogel composites in tissue regeneration. Bioengineering 2019, 6, 17. [Google Scholar] [CrossRef] [PubMed]
  92. Jahromi, M.; Razavi, S.; Seyedebrahimi, R.; Reisi, P.; Kazemi, M. Regeneration of Rat Sciatic Nerve Using PLGA Conduit Containing Rat ADSCs with Controlled Release of BDNF and Gold Nanoparticles. J. Mol. Neurosci. 2021, 71, 746–760. [Google Scholar] [CrossRef] [PubMed]
  93. Huang, L.; Yang, X.; Deng, L.; Ying, D.; Lu, A.; Zhang, L.; Yu, A.; Duan, B. Biocompatible chitin hydrogel incorporated with PEDOT nanoparticles for peripheral nerve repair. ACS Appl. Mater. Interfaces 2021, 13, 16106–16117. [Google Scholar] [CrossRef]
  94. Sun, D.; Zhuang, X.; Xiang, X.; Liu, Y.; Zhang, S.; Liu, C.; Barnes, S.; Grizzle, W.; Miller, D.; Zhang, H.-G. A Novel Nanoparticle Drug Delivery System: The Anti-inflammatory Activity of Curcumin Is Enhanced When Encapsulated in Exosomes. Mol. Ther. 2010, 18, 1606–1614. [Google Scholar] [CrossRef] [PubMed]
  95. Zhuang, X.; Xiang, X.; Grizzle, W.; Sun, D.; Zhang, S.; Axtell, R.C.; Ju, S.; Mu, J.; Zhang, L.; Steinman, L.; et al. Treatment of Brain Inflammatory Diseases by Delivering Exosome Encapsulated Anti-inflammatory Drugs From the Nasal Region to the Brain. Mol. Ther. 2011, 19, 1769–1779. [Google Scholar] [CrossRef] [PubMed]
  96. Tang, T.-T.; Lv, L.-L.; Wang, B.; Cao, J.-Y.; Feng, Y.; Li, Z.-L.; Wu, M.; Wang, F.-M.; Wen, Y.; Zhou, L.-T.; et al. Employing Macrophage-Derived Microvesicle for Kidney-Targeted Delivery of Dexamethasone: An Efficient Therapeutic Strategy against Renal Inflammation and Fibrosis. Theranostics 2019, 9, 4740–4755. [Google Scholar] [CrossRef]
  97. Lang, F.M.; Hossain, A.; Gumin, J.; Momin, E.N.; Shimizu, Y.; Ledbetter, D.; Shahar, T.; Yamashita, S.; Parker Kerrigan, B.; Fueyo, J. Mesenchymal stem cells as natural biofactories for exosomes carrying miR-124a in the treatment of gliomas. Neuro-Oncology 2018, 20, 380–390. [Google Scholar] [CrossRef]
  98. Monfared, H.; Jahangard, Y.; Nikkhah, M.; Mirnajafi-Zadeh, J.; Mowla, S.J. Potential therapeutic effects of exosomes packed with a miR-21-sponge construct in a rat model of glioblastoma. Front. Oncol. 2019, 9, 782. [Google Scholar] [CrossRef]
  99. Piffoux, M.; Silva, A.K.A.; Wilhelm, C.; Gazeau, F.; Tareste, D. Modification of Extracellular Vesicles by Fusion with Liposomes for the Design of Personalized Biogenic Drug Delivery Systems. ACS Nano 2018, 12, 6830–6842. [Google Scholar] [CrossRef]
  100. Kooijmans, S.A.A.; Fliervoet, L.A.L.; van der Meel, R.; Fens, M.H.A.M.; Heijnen, H.F.G.; van Bergen en Henegouwen, P.M.P.; Vader, P.; Schiffelers, R.M. PEGylated and targeted extracellular vesicles display enhanced cell specificity and circulation time. J. Control. Release 2016, 224, 77–85. [Google Scholar] [CrossRef]
  101. O’Loughlin, A.J.; Mäger, I.; de Jong, O.G.; Varela, M.A.; Schiffelers, R.M.; El Andaloussi, S.; Wood, M.J.A.; Vader, P. Functional Delivery of Lipid-Conjugated siRNA by Extracellular Vesicles. Mol. Ther. 2017, 25, 1580–1587. [Google Scholar] [CrossRef]
  102. de Abreu, R.C.; Ramos, C.V.; Becher, C.; Lino, M.; Jesus, C.; da Costa Martins, P.A.; Martins, P.A.; Moreno, M.J.; Fernandes, H.; Ferreira, L. Exogenous loading of miRNAs into small extracellular vesicles. J. Extracell. Vesicles 2021, 10, e12111. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, J.; Yi, K.; Zhang, Q.; Xu, H.; Zhang, X.; He, D.; Wang, F.; Xiao, X. Strong Penetration-Induced Effective Photothermal Therapy by Exosome-Mediated Black Phosphorus Quantum Dots. Small 2021, 17, 2104585. [Google Scholar] [CrossRef] [PubMed]
  104. Kim, M.S.; Haney, M.J.; Zhao, Y.; Yuan, D.; Deygen, I.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Engineering macrophage-derived exosomes for targeted paclitaxel delivery to pulmonary metastases: In vitro and in vivo evaluations. Nanomed. Nanotechnol. Biol. Med. 2018, 14, 195–204. [Google Scholar] [CrossRef] [PubMed]
  105. Haney, M.J.; Zhao, Y.; Jin, Y.S.; Li, S.M.; Bago, J.R.; Klyachko, N.L.; Kabanov, A.V.; Batrakova, E.V. Macrophage-derived extracellular vesicles as drug delivery systems for triple negative breast cancer (TNBC) therapy. J. Neuroimmune Pharmacol. 2020, 15, 487–500. [Google Scholar] [CrossRef] [PubMed]
  106. Haney, M.J.; Klyachko, N.L.; Zhao, Y.; Gupta, R.; Plotnikova, E.G.; He, Z.; Patel, T.; Piroyan, A.; Sokolsky, M.; Kabanov, A.V.; et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J. Control. Release 2015, 207, 18–30. [Google Scholar] [CrossRef]
  107. Chen, C.; Sun, M.; Wang, J.; Su, L.; Lin, J.; Yan, X. Active cargo loading into extracellular vesicles: Highlights the heterogeneous encapsulation behaviour. J. Extracell. Vesicles 2021, 10, e12163. [Google Scholar] [CrossRef]
  108. Ahmed, F.; Tamma, M.; Pathigadapa, U.; Reddanna, P.; Yenuganti, V.R. Drug loading and functional efficacy of cow, buffalo, and goat milk-derived exosomes: A comparative study. Mol. Pharm. 2022, 19, 763–774. [Google Scholar] [CrossRef]
  109. Banerjee, A.; Alves, V.; Rondão, T.; Sereno, J.; Neves, Â.; Lino, M.; Ribeiro, A.; Abrunhosa, A.J.; Ferreira, L.S. A positron-emission tomography (PET)/magnetic resonance imaging (MRI) platform to track in vivo small extracellular vesicles. Nanoscale 2019, 11, 13243–13248. [Google Scholar] [CrossRef]
  110. Lai, C.P.; Mardini, O.; Ericsson, M.; Prabhakar, S.; Maguire, C.A.; Chen, J.W.; Tannous, B.A.; Breakefield, X.O. Dynamic biodistribution of extracellular vesicles in vivo using a multimodal imaging reporter. ACS Nano 2014, 8, 483–494. [Google Scholar] [CrossRef]
  111. Hwang, D.W.; Choi, H.; Jang, S.C.; Yoo, M.Y.; Park, J.Y.; Choi, N.E.; Oh, H.J.; Ha, S.; Lee, Y.-S.; Jeong, J.M. Noninvasive imaging of radiolabeled exosome-mimetic nanovesicle using 99mTc-HMPAO. Sci. Rep. 2015, 5, 15636. [Google Scholar] [CrossRef]
  112. Kojima, R.; Bojar, D.; Rizzi, G.; Hamri, G.C.-E.; El-Baba, M.D.; Saxena, P.; Ausländer, S.; Tan, K.R.; Fussenegger, M. Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson’s disease treatment. Nat. Commun. 2018, 9, 1305. [Google Scholar] [CrossRef] [PubMed]
  113. Geng, W.; Tang, H.; Luo, S.; Lv, Y.; Liang, D.; Kang, X.; Hong, W. Exosomes from miRNA-126-modified ADSCs promotes functional recovery after stroke in rats by improving neurogenesis and suppressing microglia activation. Am. J. Transl. Res. 2019, 11, 780. [Google Scholar]
  114. Liao, X.; Gao, Z.; Xia, Y.; Zhai, W.; Pan, C.; Zhang, Y.; Yan, S.; Han, J. Micellization behavior of anionic gemini surfactants-templated manufacture of cerium oxide nanoparticles. J. Dispers. Sci. Technol. 2019, 40, 390–402. [Google Scholar] [CrossRef]
  115. Schindler, C.; Collinson, A.; Matthews, C.; Pointon, A.; Jenkinson, L.; Minter, R.R.; Vaughan, T.J.; Tigue, N.J. Exosomal delivery of doxorubicin enables rapid cell entry and enhanced in vitro potency. PLoS ONE 2019, 14, e0214545. [Google Scholar] [CrossRef] [PubMed]
  116. Shen, H.; Yao, X.; Li, H.; Li, X.; Zhang, T.; Sun, Q.; Ji, C.; Chen, G. Role of Exosomes Derived from miR-133b Modified MSCs in an Experimental Rat Model of Intracerebral Hemorrhage. J. Mol. Neurosci. 2018, 64, 421–430. [Google Scholar] [CrossRef]
  117. Sterzenbach, U.; Putz, U.; Low, L.-H.; Silke, J.; Tan, S.-S.; Howitt, J. Engineered exosomes as vehicles for biologically active proteins. Mol. Ther. 2017, 25, 1269–1278. [Google Scholar] [CrossRef]
  118. Wang, H.; Sui, H.; Zheng, Y.; Jiang, Y.; Shi, Y.; Liang, J.; Zhao, L. Curcumin-primed exosomes potently ameliorate cognitive function in AD mice by inhibiting hyperphosphorylation of the Tau protein through the AKT/GSK-3β pathway. Nanoscale 2019, 11, 7481–7496. [Google Scholar] [CrossRef]
  119. Xin, H.; Katakowski, M.; Wang, F.; Qian, J.-Y.; Liu, X.S.; Ali, M.M.; Buller, B.; Zhang, Z.G.; Chopp, M. MicroRNA-17–92 cluster in exosomes enhance neuroplasticity and functional recovery after stroke in rats. Stroke 2017, 48, 747–753. [Google Scholar] [CrossRef]
  120. Yim, N.; Ryu, S.-W.; Choi, K.; Lee, K.R.; Lee, S.; Choi, H.; Kim, J.; Shaker, M.R.; Sun, W.; Park, J.-H. Exosome engineering for efficient intracellular delivery of soluble proteins using optically reversible protein–protein interaction module. Nat. Commun. 2016, 7, 12277. [Google Scholar] [CrossRef]
  121. Zheng, Y.; He, R.; Wang, P.; Shi, Y.; Zhao, L.; Liang, J. Exosomes from LPS-stimulated macrophages induce neuroprotection and functional improvement after ischemic stroke by modulating microglial polarization. Biomater. Sci. 2019, 7, 2037–2049. [Google Scholar] [CrossRef] [PubMed]
  122. Tian, T.; Zhang, H.-X.; He, C.-P.; Fan, S.; Zhu, Y.-L.; Qi, C.; Huang, N.-P.; Xiao, Z.-D.; Lu, Z.-H.; Tannous, B.A.; et al. Surface functionalized exosomes as targeted drug delivery vehicles for cerebral ischemia therapy. Biomaterials 2018, 150, 137–149. [Google Scholar] [CrossRef] [PubMed]
  123. Jia, G.; Han, Y.; An, Y.; Ding, Y.; He, C.; Wang, X.; Tang, Q. NRP-1 targeted and cargo-loaded exosomes facilitate simultaneous imaging and therapy of glioma in vitro and in vivo. Biomaterials 2018, 178, 302–316. [Google Scholar] [CrossRef] [PubMed]
  124. Khongkow, M.; Yata, T.; Boonrungsiman, S.; Ruktanonchai, U.R.; Graham, D.; Namdee, K. Surface modification of gold nanoparticles with neuron-targeted exosome for enhanced blood–brain barrier penetration. Sci. Rep. 2019, 9, 8278. [Google Scholar] [CrossRef]
  125. Kooijmans, S.A.; Aleza, C.G.; Roffler, S.R.; van Solinge, W.W.; Vader, P.; Schiffelers, R.M. Display of GPI-anchored anti-EGFR nanobodies on extracellular vesicles promotes tumour cell targeting. J. Extracell. Vesicles 2016, 5, 31053. [Google Scholar] [CrossRef] [PubMed]
  126. Lee, J.; Lee, H.; Goh, U.; Kim, J.; Jeong, M.; Lee, J.; Park, J.-H. Cellular engineering with membrane fusogenic liposomes to produce functionalized extracellular vesicles. ACS Appl. Mater. Interfaces 2016, 8, 6790–6795. [Google Scholar] [CrossRef]
  127. Nakase, I.; Futaki, S. Combined treatment with a pH-sensitive fusogenic peptide and cationic lipids achieves enhanced cytosolic delivery of exosomes. Sci. Rep. 2015, 5, 10112. [Google Scholar] [CrossRef]
  128. Qi, H.; Liu, C.; Long, L.; Ren, Y.; Zhang, S.; Chang, X.; Qian, X.; Jia, H.; Zhao, J.; Sun, J. Blood exosomes endowed with magnetic and targeting properties for cancer therapy. ACS Nano 2016, 10, 3323–3333. [Google Scholar] [CrossRef]
  129. Smyth, T.; Petrova, K.; Payton, N.M.; Persaud, I.; Redzic, J.S.; Graner, M.W.; Smith-Jones, P.; Anchordoquy, T.J. Surface functionalization of exosomes using click chemistry. Bioconjugate Chem. 2014, 25, 1777–1784. [Google Scholar] [CrossRef]
  130. Zhang, H.; Wu, J.; Wu, J.; Fan, Q.; Zhou, J.; Wu, J.; Liu, S.; Zang, J.; Ye, J.; Xiao, M. Exosome-mediated targeted delivery of miR-210 for angiogenic therapy after cerebral ischemia in mice. J. Nanobiotechnol. 2019, 17, 29. [Google Scholar] [CrossRef]
  131. Wang, M.; Altinoglu, S.; Takeda, Y.S.; Xu, Q. Integrating Protein Engineering and Bioorthogonal Click Conjugation for Extracellular Vesicle Modulation and Intracellular Delivery. PLoS ONE 2015, 10, e0141860. [Google Scholar] [CrossRef]
  132. Yang, J.; Zhang, X.; Chen, X.; Wang, L.; Yang, G. Exosome mediated delivery of miR-124 promotes neurogenesis after ischemia. Mol. Ther.-Nucleic Acids 2017, 7, 278–287. [Google Scholar] [CrossRef] [PubMed]
  133. Silva, A.K.A.; Luciani, N.; Gazeau, F.; Aubertin, K.; Bonneau, S.; Chauvierre, C.; Letourneur, D.; Wilhelm, C. Combining magnetic nanoparticles with cell derived microvesicles for drug loading and targeting. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 645–655. [Google Scholar] [CrossRef]
  134. Zhang, G.; Chen, L.; Guo, X.; Wang, H.; Chen, W.; Wu, G.; Gu, B.; Miao, W.; Kong, J.; Jin, X. Comparative analysis of microRNA expression profiles of exosomes derived from normal and hypoxic preconditioning human neural stem cells by next generation sequencing. J. Biomed. Nanotechnol. 2018, 14, 1075–1089. [Google Scholar] [CrossRef] [PubMed]
  135. Wang, J.; Chen, S.; Zhang, W.; Chen, Y.; Bihl, J.C. Exosomes from miRNA-126-modified endothelial progenitor cells alleviate brain injury and promote functional recovery after stroke. CNS Neurosci. Ther. 2020, 26, 1255–1265. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, C.; Börger, V.; Sardari, M.; Murke, F.; Skuljec, J.; Pul, R.; Hagemann, N.; Dzyubenko, E.; Dittrich, R.; Gregorius, J. Mesenchymal Stromal Cell–Derived Small Extracellular Vesicles Induce Ischemic Neuroprotection by Modulating Leukocytes and Specifically Neutrophils. Stroke 2020, 51, 1825–1834. [Google Scholar] [CrossRef] [PubMed]
  137. Laso-García, F.; Casado-Fernández, L.; Piniella, D.; Gómez-de Frutos, M.C.; Arizaga-Echebarria, J.K.; Pérez-Mato, M.; Alonso-López, E.; Otero-Ortega, L.; Bravo, S.B.; del Pilar Chantada-Vázquez, M. Circulating extracellular vesicles promote recovery in a preclinical model of intracerebral hemorrhage. Mol. Ther.-Nucleic Acids 2023, 32, 247–262. [Google Scholar] [CrossRef] [PubMed]
  138. Ahn, S.Y.; Sung, D.K.; Kim, Y.E.; Sung, S.; Chang, Y.S.; Park, W.S. Brain-derived neurotropic factor mediates neuroprotection of mesenchymal stem cell-derived extracellular vesicles against severe intraventricular hemorrhage in newborn rats. Stem Cells Transl. Med. 2021, 10, 374–384. [Google Scholar] [CrossRef]
  139. Gao, X.; Xiong, Y.; Li, Q.; Han, M.; Shan, D.; Yang, G.; Zhang, S.; Xin, D.; Zhao, R.; Wang, Z.; et al. Extracellular vesicle-mediated transfer of miR-21-5p from mesenchymal stromal cells to neurons alleviates early brain injury to improve cognitive function via the PTEN/Akt pathway after subarachnoid hemorrhage. Cell Death Dis. 2020, 11, 363. [Google Scholar] [CrossRef]
  140. Ding, H.; Jia, Y.; Lv, H.; Chang, W.; Liu, F.; Wang, D. Extracellular vesicles derived from bone marrow mesenchymal stem cells alleviate neuroinflammation after diabetic intracerebral hemorrhage via the miR-183-5p/PDCD4/NLRP3 pathway. J. Endocrinol. Investig. 2021, 44, 2685–2698. [Google Scholar] [CrossRef]
  141. Wang, J.; Chen, S.; Meghana Yerrapragada, S.; Zhang, W.; Bihl, J.C. Therapeutic effects of exosomes from angiotensin-converting enzyme 2 -overexpressed endothelial progenitor cells on intracerebral hemorrhagic stroke. Brain Hemorrhages 2021, 2, 57–62. [Google Scholar] [CrossRef]
  142. Liao, K.; Niu, F.; Dagur, R.S.; He, M.; Tian, C.; Hu, G. Intranasal delivery of lincRNA-Cox2 siRNA loaded extracellular vesicles decreases lipopolysaccharide-induced microglial proliferation in mice. J. Neuroimmune Pharmacol. 2020, 15, 390–399. [Google Scholar] [CrossRef] [PubMed]
  143. Xin, H.; Li, Y.; Chopp, M. Exosomes/miRNAs as mediating cell-based therapy of stroke. Front. Cell. Neurosci. 2014, 8, 377. [Google Scholar] [CrossRef]
  144. Liu, Y.; Li, D.; Liu, Z.; Zhou, Y.; Chu, D.; Li, X.; Jiang, X.; Hou, D.; Chen, X.; Chen, Y. Targeted exosome-mediated delivery of opioid receptor Mu siRNA for the treatment of morphine relapse. Sci. Rep. 2015, 5, 17543. [Google Scholar] [CrossRef]
  145. Ahmed, W.; Khan, A.; Sundar, W.H.; Naseem, H.; Chen, W.; Feng, J.; Durrani, S.; Chen, L. Neurological diseases caused by coronavirus infection of the respiratory airways. Brain Sci. Adv. 2020, 6, 324–343. [Google Scholar] [CrossRef]
  146. Qi, Y.; Guo, L.; Jiang, Y.; Shi, Y.; Sui, H.; Zhao, L. Brain delivery of quercetin-loaded exosomes improved cognitive function in AD mice by inhibiting phosphorylated tau-mediated neurofibrillary tangles. Drug Deliv. 2020, 27, 745–755. [Google Scholar] [CrossRef] [PubMed]
  147. Zhu, Q.; Ling, X.; Yang, Y.; Zhang, J.; Li, Q.; Niu, X.; Hu, G.; Chen, B.; Li, H.; Wang, Y.; et al. Embryonic stem cells-derived exosomes endowed with targeting properties as chemotherapeutics delivery vehicles for glioblastoma therapy. Adv. Sci. 2019, 6, 1801899. [Google Scholar] [CrossRef]
  148. Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome delivered anticancer drugs across the blood-brain barrier for brain cancer therapy in Danio rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef]
  149. Hu, G.; Liao, K.; Niu, F.; Yang, L.; Dallon, B.W.; Callen, S.; Tian, C.; Shu, J.; Cui, J.; Sun, Z.; et al. Astrocyte EV-Induced lincRNA-Cox2 Regulates Microglial Phagocytosis: Implications for Morphine-Mediated Neurodegeneration. Mol. Ther.-Nucleic Acids 2018, 13, 450–463. [Google Scholar] [CrossRef]
  150. Qu, M.; Lin, Q.; Huang, L.; Fu, Y.; Wang, L.; He, S.; Fu, Y.; Yang, S.; Zhang, Z.; Zhang, L.; et al. Dopamine-loaded blood exosomes targeted to brain for better treatment of Parkinson’s disease. J. Control. Release 2018, 287, 156–166. [Google Scholar] [CrossRef]
  151. Liu, J.; Duan, W.; Deng, Y.; Zhang, Q.; Li, R.; Long, J.; Ahmed, W.; Gu, C.; Qiu, Y.; Cai, H. New Insights into Molecular Mechanisms Underlying Neurodegenerative Disorders. J. Integr. Neurosci. 2023, 22, 58. [Google Scholar] [CrossRef] [PubMed]
  152. Yong, T.; Zhang, X.; Bie, N.; Zhang, H.; Zhang, X.; Li, F.; Hakeem, A.; Hu, J.; Gan, L.; Santos, H.A. Tumor exosome-based nanoparticles are efficient drug carriers for chemotherapy. Nat. Commun. 2019, 10, 3838. [Google Scholar] [CrossRef] [PubMed]
  153. Zhang, Z.G.; Buller, B.; Chopp, M. Exosomes—Beyond stem cells for restorative therapy in stroke and neurological injury. Nat. Rev. Neurol. 2019, 15, 193–203. [Google Scholar] [CrossRef] [PubMed]
  154. Xu, L.; Yang, X.; Gao, H.; Wang, X.; Zhou, B.; Li, Y.; Li, L.; Guo, X.; Ren, L. Clinical efficacy and safety analysis of argatroban and alteplase treatment regimens for acute cerebral infarction. J. Neurorestoratology 2022, 10, 100017. [Google Scholar] [CrossRef]
  155. Kim, H.Y.; Kim, T.J.; Kang, L.; Kim, Y.-J.; Kang, M.K.; Kim, J.; Ryu, J.H.; Hyeon, T.; Yoon, B.-W.; Ko, S.-B. Mesenchymal stem cell-derived magnetic extracellular nanovesicles for targeting and treatment of ischemic stroke. Biomaterials 2020, 243, 119942. [Google Scholar] [CrossRef] [PubMed]
  156. Zhu, Z.; Kalyan, B.S.; Chen, L. Therapeutic potential role of exosomes for ischemic stroke. Brain Sci. Adv. 2019, 5, 128–143. [Google Scholar] [CrossRef]
  157. Feng, J.; Zhang, Y.; Zhu, Z.; Gu, C.; Waqas, A.; Chen, L. Emerging exosomes and exosomal MiRNAs in spinal cord injury. Front. Cell Dev. Biol. 2021, 9, 703989. [Google Scholar] [CrossRef]
  158. Xu, T.; Ma, Y.; Yuan, Q.; Hu, H.; Hu, X.; Qian, Z.; Rolle, J.K.; Gu, Y.; Li, S. Enhanced ferroptosis by oxygen-boosted phototherapy based on a 2-in-1 nanoplatform of ferrous hemoglobin for tumor synergistic therapy. ACS Nano 2020, 14, 3414–3425. [Google Scholar] [CrossRef]
  159. Gu, C.; Zhang, Q.; Li, Y.; Li, R.; Feng, J.; Chen, W.; Ahmed, W.; Soufiany, I.; Huang, S.; Long, J. The PI3K/AKT Pathway—The Potential Key Mechanisms of Traditional Chinese Medicine for Stroke. Front. Med. 2022, 9, 900809. [Google Scholar] [CrossRef]
  160. Moura, R.P.; Sousa, F.; Almeida, A.; Pinto, S.; Sarmento, B. Theranostic biomaterials for regulation of the blood–brain barrier. In Theranostic Bionanomaterials; Elsevier: Amsterdam, The Netherlands, 2019; pp. 303–319. [Google Scholar]
  161. Yang, D.; Shao, J.; Hu, R.; Chen, H.; Xie, P.; Liu, C. Angiotensin II promotes the anticoagulant effects of rivaroxaban via angiotensin type 2 receptor signaling in mice. Sci. Rep. 2017, 7, 369. [Google Scholar] [CrossRef]
  162. Doll, D.N.; Barr, T.L.; Simpkins, J.W. Cytokines: Their role in stroke and potential use as biomarkers and therapeutic targets. Aging Dis. 2014, 5, 294. [Google Scholar] [CrossRef]
  163. Klimiec-Moskal, E.; Piechota, M.; Pera, J.; Weglarczyk, K.; Slowik, A.; Siedlar, M.; Dziedzic, T. The specific ex vivo released cytokine profile is associated with ischemic stroke outcome and improves its prediction. J. Neuroinflamm. 2020, 17, 7. [Google Scholar] [CrossRef] [PubMed]
  164. Zhang, G.; Zhu, Z.; Wang, H.; Yu, Y.; Chen, W.; Waqas, A.; Wang, Y.; Chen, L. Exosomes derived from human neural stem cells stimulated by interferon gamma improve therapeutic ability in ischemic stroke model. J. Adv. Res. 2020, 24, 435–445. [Google Scholar] [CrossRef] [PubMed]
  165. Chaudhuri, A.D.; Dastgheyb, R.M.; Yoo, S.-W.; Trout, A.; Talbot, C.C., Jr.; Hao, H.; Witwer, K.W.; Haughey, N.J. TNFα and IL-1β modify the miRNA cargo of astrocyte shed extracellular vesicles to regulate neurotrophic signaling in neurons. Cell Death Dis. 2018, 9, 363. [Google Scholar] [CrossRef] [PubMed]
  166. Dickens, A.M.; Tovar-y-Romo, L.B.; Yoo, S.-W.; Trout, A.L.; Bae, M.; Kanmogne, M.; Megra, B.; Williams, D.W.; Witwer, K.W.; Gacias, M. Astrocyte-shed extracellular vesicles regulate the peripheral leukocyte response to inflammatory brain lesions. Sci. Signal. 2017, 10, eaai7696. [Google Scholar] [CrossRef]
  167. Tian, Y.; Zhu, P.; Liu, S.; Jin, Z.; Li, D.; Zhao, H.; Zhu, X.; Shu, C.; Yan, D.; Dong, Z. IL-4-polarized BV2 microglia cells promote angiogenesis by secreting exosomes. Adv. Clin. Exp. Med. 2019, 28, 421–430. [Google Scholar] [CrossRef]
  168. Zhu, Z.H.; Jia, F.; Ahmed, W.; Zhang, G.L.; Wang, H.; Lin, C.Q.; Chen, W.H.; Chen, L.K. Neural stem cell-derived exosome as a nano-sized carrier for BDNF delivery to a rat model of ischemic stroke. Neural Regen. Res. 2023, 18, 404–409. [Google Scholar] [CrossRef]
  169. Attia, Z.I.; Kapa, S.; Lopez-Jimenez, F.; McKie, P.M.; Ladewig, D.J.; Satam, G.; Pellikka, P.A.; Enriquez-Sarano, M.; Noseworthy, P.A.; Munger, T.M. Screening for cardiac contractile dysfunction using an artificial intelligence–enabled electrocardiogram. Nat. Med. 2019, 25, 70–74. [Google Scholar] [CrossRef]
  170. Su, Y.-L.; Kuo, L.-W.; Hsu, C.-H.; Chiang, C.-S.; Lu, Y.-J.; Chang, S.-J.; Hu, S.-H. Rabies virus glycoprotein-amplified hierarchical targeted hybrids capable of magneto-electric penetration delivery to orthotopic brain tumor. J. Control. Release 2020, 321, 159–173. [Google Scholar] [CrossRef]
  171. Chivero, E.T.; Liao, K.; Niu, F.; Tripathi, A.; Tian, C.; Buch, S.; Hu, G. Engineered extracellular vesicles loaded with miR-124 attenuate cocaine-mediated activation of microglia. Front. Cell Dev. Biol. 2020, 8, 573. [Google Scholar] [CrossRef]
  172. Yang, L.; Han, B.; Zhang, Z.; Wang, S.; Bai, Y.; Zhang, Y.; Tang, Y.; Du, L.; Xu, L.; Wu, F.; et al. Extracellular Vesicle-Mediated Delivery of Circular RNA SCMH1 Promotes Functional Recovery in Rodent and Nonhuman Primate Ischemic Stroke Models. Circulation 2020, 142, 556–574. [Google Scholar] [CrossRef] [PubMed]
  173. Lindholm, P.; Saarma, M. Novel CDNF/MANF family of neurotrophic factors. Dev. Neurobiol. 2010, 70, 360–371. [Google Scholar] [CrossRef] [PubMed]
  174. Houlton, J.; Abumaria, N.; Hinkley, S.F.; Clarkson, A.N. Therapeutic potential of neurotrophins for repair after brain injury: A helping hand from biomaterials. Front. Neurosci. 2019, 13, 790. [Google Scholar] [CrossRef]
  175. Abe, K. Therapeutic potential of neurotrophic factors and neural stem cells against ischemic brain injury. J. Cereb. Blood Flow Metab. 2000, 20, 1393–1408. [Google Scholar] [CrossRef] [PubMed]
  176. Khan, H.; Pan, J.-J.; Li, Y.; Zhang, Z.; Yang, G.-Y. Native and bioengineered exosomes for ischemic stroke therapy. Front. Cell Dev. Biol. 2021, 9, 619565. [Google Scholar] [CrossRef]
  177. Ramos-Cejudo, J.; Gutiérrez-Fernández, M.; Otero-Ortega, L.; Rodríguez-Frutos, B.; Fuentes, B.; Vallejo-Cremades, M.T.; Hernanz, T.N.; Cerdán, S.; Díez-Tejedor, E. Brain-derived neurotrophic factor administration mediated oligodendrocyte differentiation and myelin formation in subcortical ischemic stroke. Stroke 2015, 46, 221–228. [Google Scholar] [CrossRef]
  178. Lin, C.-Q.; Chen, L.-K. Cerebral dopamine neurotrophic factor promotes the proliferation and differentiation of neural stem cells in hypoxic environments. Neural Regen. Res. 2020, 15, 2057. [Google Scholar]
  179. Donoso-Quezada, J.; Ayala-Mar, S.; González-Valdez, J. State-of-the-art exosome loading and functionalization techniques for enhanced therapeutics: A review. Crit. Rev. Biotechnol. 2020, 40, 804–820. [Google Scholar] [CrossRef]
  180. Zhao, H.; Li, Y.; Chen, L.; Shen, C.; Xiao, Z.; Xu, R.; Wang, J.; Luo, Y. HucMSCs-derived miR-206-knockdown exosomes contribute to neuroprotection in subarachnoid hemorrhage induced early brain injury by targeting BDNF. Neuroscience 2019, 417, 11–23. [Google Scholar] [CrossRef]
  181. Izadpanah, M.; Seddigh, A.; Ebrahimi Barough, S.; Fazeli, S.A.S.; Ai, J. Potential of Extracellular Vesicles in Neurodegenerative Diseases: Diagnostic and Therapeutic Indications. J. Mol. Neurosci. 2018, 66, 172–179. [Google Scholar] [CrossRef]
  182. Morad, G.; Carman, C.V.; Hagedorn, E.J.; Perlin, J.R.; Zon, L.I.; Mustafaoglu, N.; Park, T.-E.; Ingber, D.E.; Daisy, C.C.; Moses, M.A. Tumor-derived extracellular vesicles breach the intact blood–brain barrier via transcytosis. ACS Nano 2019, 13, 13853–13865. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Composition and Structure of EVs. The structure of EVs consists of a phospholipid bilayer that encloses proteins (membrane protein and cargo protein) and nucleic acids. Membrane proteins encompass a variety of molecules, such as tetraspanins (including CD9, CD63, and CD81, among others), adhesion molecules (such as integrins, EpCAM, and Ephrin), the major histocompatibility complex (MHC), and receptors. Nucleic acids encompass DNA and RNA, which consist of various types of RNA molecules, such as messenger RNA (mRNA), microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA). The phospholipid bilayer confers protection to the contents enclosed within. Created with BioRender.com (https://app.biorender.com/illustrations/649b3cd0422a45d3d7cc3c29, accessed date: 7 July 2023).
Figure 1. Composition and Structure of EVs. The structure of EVs consists of a phospholipid bilayer that encloses proteins (membrane protein and cargo protein) and nucleic acids. Membrane proteins encompass a variety of molecules, such as tetraspanins (including CD9, CD63, and CD81, among others), adhesion molecules (such as integrins, EpCAM, and Ephrin), the major histocompatibility complex (MHC), and receptors. Nucleic acids encompass DNA and RNA, which consist of various types of RNA molecules, such as messenger RNA (mRNA), microRNA (miRNA), long non-coding RNA (lncRNA), and circular RNA (circRNA). The phospholipid bilayer confers protection to the contents enclosed within. Created with BioRender.com (https://app.biorender.com/illustrations/649b3cd0422a45d3d7cc3c29, accessed date: 7 July 2023).
Pharmaceutics 15 02173 g001
Figure 2. Schematic of EV Subtypes. (A) Exosomes and their formation. (B) Microvesicles and their formation. (C) Apoptotic bodies and their formation. Created with BioRender.com (https://app.biorender.com/illustrations/64d0be423f71706d207a7881, accessed date: 7 August 2023).
Figure 2. Schematic of EV Subtypes. (A) Exosomes and their formation. (B) Microvesicles and their formation. (C) Apoptotic bodies and their formation. Created with BioRender.com (https://app.biorender.com/illustrations/64d0be423f71706d207a7881, accessed date: 7 August 2023).
Pharmaceutics 15 02173 g002
Figure 3. Isolation and quantification techniques for EVs. The diagram illustrates frequently employed methods for analyzing EVs. EVs can be quantified in tissue homogenates and natural fluids, such as urine, saliva, and blood. The isolation of plasma or serum from blood is utilized in this context as an illustrative case. Nanoflow cytometry enables direct labeling and quantifying EVs in various fluid samples. In contrast, EVs have the potential to be separated and subsequently utilized for further analysis. Immunoprecipitation is a technique that can be employed to enhance the specificity of EV populations. EVs can be observed using electron microscopy (EM) or alternative high-resolution microscopy methodologies. Lipidomics and proteomics methodologies can also be utilized to analyze and describe the composition of EVs populations. Ultimately, EV concentration and size measurement can now be achieved through dynamic light scattering and nanoparticle tracking analysis. Additionally, EV products can be measured by employing susceptible protein or RNA assays. Created with BioRender.com (https://app.biorender.com/illustrations/649b163862468f1db106b519, Accessed date: 7 August 2023).
Figure 3. Isolation and quantification techniques for EVs. The diagram illustrates frequently employed methods for analyzing EVs. EVs can be quantified in tissue homogenates and natural fluids, such as urine, saliva, and blood. The isolation of plasma or serum from blood is utilized in this context as an illustrative case. Nanoflow cytometry enables direct labeling and quantifying EVs in various fluid samples. In contrast, EVs have the potential to be separated and subsequently utilized for further analysis. Immunoprecipitation is a technique that can be employed to enhance the specificity of EV populations. EVs can be observed using electron microscopy (EM) or alternative high-resolution microscopy methodologies. Lipidomics and proteomics methodologies can also be utilized to analyze and describe the composition of EVs populations. Ultimately, EV concentration and size measurement can now be achieved through dynamic light scattering and nanoparticle tracking analysis. Additionally, EV products can be measured by employing susceptible protein or RNA assays. Created with BioRender.com (https://app.biorender.com/illustrations/649b163862468f1db106b519, Accessed date: 7 August 2023).
Pharmaceutics 15 02173 g003
Figure 4. Choices and obstacles while using EVs. The application of EVs originating from diverse cellular origins is commonly observed in managing stroke. The properties of EVs associated with each cell type may have varying levels of tropism for brain vasculature or neuronal cells, which could impact their ability to target the brain effectively. However, a complete evaluation of these properties has yet to be conducted. Crossing the blood–brain barrier (BBB) poses a significant challenge. The method of administration affects the biodistribution and clearance of EVs and can also impact the effect’s nature, i.e., whether it is localized or systematic. Lastly, the dosing schedule can be single or repeated, affecting accumulation and efficacy. Created with BioRender.com (https://app.biorender.com/illustrations/647df1821b2f09af295c29aa, accessed Date: 5 June 2023).
Figure 4. Choices and obstacles while using EVs. The application of EVs originating from diverse cellular origins is commonly observed in managing stroke. The properties of EVs associated with each cell type may have varying levels of tropism for brain vasculature or neuronal cells, which could impact their ability to target the brain effectively. However, a complete evaluation of these properties has yet to be conducted. Crossing the blood–brain barrier (BBB) poses a significant challenge. The method of administration affects the biodistribution and clearance of EVs and can also impact the effect’s nature, i.e., whether it is localized or systematic. Lastly, the dosing schedule can be single or repeated, affecting accumulation and efficacy. Created with BioRender.com (https://app.biorender.com/illustrations/647df1821b2f09af295c29aa, accessed Date: 5 June 2023).
Pharmaceutics 15 02173 g004
Figure 5. The role of natural EVs in the pathophysiology of stroke. The management of stroke has shown benefits through direct local effects in the brain, such as neuroprotection, neurogenesis, angiogenesis, antioxidant and anti-inflammatory properties, and systemic effects by modulating peripheral immune system responses. These effects may create a favorable environment for cerebral regeneration. Created with BioRender.com (https://app.biorender.com/illustrations/647df0bfe74d4f82a5bc4ade, accessed: 11 June 2023).
Figure 5. The role of natural EVs in the pathophysiology of stroke. The management of stroke has shown benefits through direct local effects in the brain, such as neuroprotection, neurogenesis, angiogenesis, antioxidant and anti-inflammatory properties, and systemic effects by modulating peripheral immune system responses. These effects may create a favorable environment for cerebral regeneration. Created with BioRender.com (https://app.biorender.com/illustrations/647df0bfe74d4f82a5bc4ade, accessed: 11 June 2023).
Pharmaceutics 15 02173 g005
Figure 6. Natural EV modes of action in stroke. The transfer of MiR-133b through EVs originating from mesenchymal stem cells (MSCs) has been found to facilitate the growth of neurites. This effect is achieved through the targeting of the converting protein RhoA. Additionally, miR-124 is linked to increased neuronal viability by targeting USP-14, a ubiquitin-specific protease. Systemically, it has been demonstrated that EVs from NSC reduce pro-inflammatory Th17 cells while improving immunosuppressive Treg cells. Created with BioRender.com (https://app.biorender.com/illustrations/647ca600a95ee7a5e7fd6757, accessed: 11 June 2023).
Figure 6. Natural EV modes of action in stroke. The transfer of MiR-133b through EVs originating from mesenchymal stem cells (MSCs) has been found to facilitate the growth of neurites. This effect is achieved through the targeting of the converting protein RhoA. Additionally, miR-124 is linked to increased neuronal viability by targeting USP-14, a ubiquitin-specific protease. Systemically, it has been demonstrated that EVs from NSC reduce pro-inflammatory Th17 cells while improving immunosuppressive Treg cells. Created with BioRender.com (https://app.biorender.com/illustrations/647ca600a95ee7a5e7fd6757, accessed: 11 June 2023).
Pharmaceutics 15 02173 g006
Figure 7. EV/NP. Transport Neural Remodeling. Poly(3,4-ethylenedioxythiophene) modified with tetrapeptide was administered utilizing a biocompatible chitin scaffold. In an in vivo model, myelin thickness increased, indicating nerve regeneration. At the injury’s site, Schwann cell adhesiveness and angiogenesis increased. Created with BioRender.com (https://app.biorender.com/illustrations/64a80816d2fc5a3659e189a9, accessed: 7 July 2023).
Figure 7. EV/NP. Transport Neural Remodeling. Poly(3,4-ethylenedioxythiophene) modified with tetrapeptide was administered utilizing a biocompatible chitin scaffold. In an in vivo model, myelin thickness increased, indicating nerve regeneration. At the injury’s site, Schwann cell adhesiveness and angiogenesis increased. Created with BioRender.com (https://app.biorender.com/illustrations/64a80816d2fc5a3659e189a9, accessed: 7 July 2023).
Pharmaceutics 15 02173 g007
Figure 8. Engineering strategies for modifying EV content. Cellular engineering techniques can regulate EVs, including altering primitive cells or directly loading them via various post-isolation methods. (A) Cell engineering uses genetic manipulation techniques, such as plasmid transfection or enriching cells with miRNAs or small compounds, to load a parent cell indirectly. (B) Electroporation, sonication, freeze–thaw cycles, and chemical agents modulate isolated EVs post-isolation. Therapeutic substances and loading efficiency determine the optimum EV modulation strategy. EVs cargo manipulation can treat stroke by acting on cargo type. (C) Proteins. (D) Small molecules. (E) miRNAs. Created with BioRender.com (https://app.biorender.com/illustrations/647dfe244f0c62bb59ac8cb8, accessed: 6 June 2023).
Figure 8. Engineering strategies for modifying EV content. Cellular engineering techniques can regulate EVs, including altering primitive cells or directly loading them via various post-isolation methods. (A) Cell engineering uses genetic manipulation techniques, such as plasmid transfection or enriching cells with miRNAs or small compounds, to load a parent cell indirectly. (B) Electroporation, sonication, freeze–thaw cycles, and chemical agents modulate isolated EVs post-isolation. Therapeutic substances and loading efficiency determine the optimum EV modulation strategy. EVs cargo manipulation can treat stroke by acting on cargo type. (C) Proteins. (D) Small molecules. (E) miRNAs. Created with BioRender.com (https://app.biorender.com/illustrations/647dfe244f0c62bb59ac8cb8, accessed: 6 June 2023).
Pharmaceutics 15 02173 g008
Figure 9. Surface modulation of EVs. The modulation of EV surfaces can be accomplished by genetically modifying the cells that produce them. (A) Protein plasmids or (B) Protein-residues (C) directly conjugated to lipids that are then integrated into EV membranes. (D) Bio-orthogonal chemistry identifies extracellular vesicle functional groups. Created with BioRender.com (https://app.biorender.com/illustrations/6480cf6ae6947f82704ea72e, accessed: 11 June 2023).
Figure 9. Surface modulation of EVs. The modulation of EV surfaces can be accomplished by genetically modifying the cells that produce them. (A) Protein plasmids or (B) Protein-residues (C) directly conjugated to lipids that are then integrated into EV membranes. (D) Bio-orthogonal chemistry identifies extracellular vesicle functional groups. Created with BioRender.com (https://app.biorender.com/illustrations/6480cf6ae6947f82704ea72e, accessed: 11 June 2023).
Pharmaceutics 15 02173 g009
Table 1. Natural EVs for the treatment of stroke.
Table 1. Natural EVs for the treatment of stroke.
DiseaseModelEV SourceRouteDoseOutcomeReference
StrokeMCAO in mouseHuman MSCsIVMultiple administrations↑ Neurogenesis
↑ Angiogenesis
[53]
M2 MicrogliaIV100 μg↑ Neuron protection
↓ Volume of infarction
[54]
Mouse NSCsIV100 μg↑ Availability of astrocytes
↓ Volume of infarction
[27]
Mouse NSCs and MSCsRO1–100 μg (multiple administrations)↓ Impaired motor coordination
↑ Neuro-regeneration
[55,56]
TE-MCAO in mouseHuman NSCsIV2.7 × 1011 EVs/kg (multiple administrations)↓ Cerebral atrophy
↑ Motor recovery
[57,58]
MCAO in ratPorcine MSCsIV100 μg↑ Functional recovery
↓ Volume of infarct
↑ Angiogenesis
[59]
Human MSCsIA200 μg/kg↑ Functional recovery
↓ Volume of infarct
↑ Angiogenesis
[60]
Rat NSCsICV30 μg↑ Neural protection
↓ Microgliosis
↓ Size of infarct
↓ Behavioral deficits
[61,62]
Rat MSCsICV100 μg↓ Size of infarct
↑ Functional recovery
[63]
Rat MSCsIV100 μg↑ Neuron transformation[24]
MSCs—Mesenchymal stem cells; IV—intravenous injection; MCAO—middle cerebral artery occlusion; NSCs—neural stem cells; TE-MCAO—thromboembolic middle cerebral artery occlusion; RO—retro-orbital; ICV—intracerebroventricularly. ↑ Increase, ↓ Decrease.
Table 2. Methods for loading EVs with drugs.
Table 2. Methods for loading EVs with drugs.
Loading StrategiesLoading MethodsAdvantagesDisadvantagesReference
Pre-loadingCo-incubation1. Simple
2. Cost-effective
3. EV-friendly
1. Low encapsulation efficiency
2. Strict cargo selection
[94,95,96]
TransfectionTarget molecule overexpression1. Time-consuming
2. Highly dependent on cell viability
3. Potential toxicity and genetic changes
[97,98]
Post-loadingCo-incubation1. Easy operation
2. No extra equipment is required
3. Minimal destruction to EVs
1. Low loading efficiency
2. Limited variety
[99,100,101]
Electroporation1. Effective loading efficiency
2. Loading of large biomolecules
1. Affect EVs integrity
2. Risk of EVs aggregation
3. Heat can cause damage
[102,103]
SonicationHigh loading efficiency1. EVs membrane degradation
2. EV aggregation risk
[104,105]
Freeze–thawing cycle1. Cost-effective
2. Applicable for most cargoes
1. Low loading efficiency
2. EVs membrane damage
3. EVs aggregation risk
[106,107]
Surfactant administration1. Affordable
2. Applicable for most cargoes
EVs surface potential and functionality may be altered[76,108]
Table 3. Techniques for modifying EVs cargo in stroke.
Table 3. Techniques for modifying EVs cargo in stroke.
Origin of EVsMethodModelResultReference
Rat MSCsmiR-17-92 cluster overexpressionMCAO rat model↑ Neurogenesis
↑ Neurological function
[119]
Rat MSCsmiR-133b overexpressionMCAO rat model↑ Neuroprotection[116]
Human ADSCsmiR-126 overexpressionRat MCAO↑ Neurogenesis
↑ Angiogenesis
↓ Inflammation
[113,134]
Mouse EPCsmiR-126 overexpressionMouse MCAO↓ Infarct size
↑ Neurogenesis
↑ Angiogenesis
[135]
Mouse MSCsDiffusion of curcumin-loaded EVsMouse MCAO model↓ Inflammation
↓ Neuronal apoptosis
[122]
Human MSCsDiffusion of leucocyte-loaded EVsMouse MCAO model↓ Brain leukocyte infiltration
↑ Neuroprotection
[136]
Rat-blood-derived EVsPCSK9
overexpression
Mouse ICH model↑ Neuroprotection
↑ Myelination
↑ Angiogenesis
[137]
Human umbilical cord blood (UCB)–MSC-derived EVsDiffusion of
BDNF-Loaded EVs
Rat IVH
model
↑ Neuroprotection
↓ Inflammatory response/
Apoptosis/
↑ Myelination and neurogenesis
[138]
Bone-marrow–MSCs derived EVsmiR-21-5p
overexpression
Rat SAH model↓ Neuronal apoptosis
↑ Neuroprotection
[139]
BMSC-derived EVsmiR-183-5p
overexpression
Rat model of db-ICH↓ Neuroinflammation
↓ Neurological deficit
[140]
EVs from angiotensin-converting enzyme 2 (ACE 2)Endothelial progenitor cells overexpressionMouse ICH model↓ Decreased hemorrhage volume
↓ Brain edema
Improved Neurological Deficit Score (NDS)
[141]
MSCs—Mesenchymal stem cells; ADSCs—adipose-derived stem cells; EPCs—endothelial progenitor cells; EVs—extracellular vesicles; MCAO—middle cerebral artery occlusion; ICH—intracerebral hemorrhage; IVH—intraventricular hemorrhage; SAH—subarachnoid hemorrhage; BMSCs—bone marrow mesenchymal stem cells; db-ICH—diabetic intracerebral hemorrhage. ↑ Increase, ↓ Decrease.
Table 4. EV cargo modification in other neurological conditions.
Table 4. EV cargo modification in other neurological conditions.
Origin of EVsMethodModelResultReference
Mouse embryonic fibroblastsCre recombinase enzyme overexpressionTransgenic mouse modelIntranasal transport of brain-active proteins[117]
Human HEK-293 TOverexpression of the catalase enzymePD mouse model↓ Neuronal inflammation[112]
Mouse astrocytesTransfection with lincRNA-Cox2-siRNAIn vitro/in vivo lincRNA-Cox2 knockout model Intranasal↓ Expression of lincRNA-Cox2;
LPS-induced microglial proliferation
[114]
Human astrocytesTransfection with lincRNA-Cox2-siRNAIn vitro/in vivo lincRNA-Cox2 knockout model IntranasalMicroglial phagocytic activity restored[149]
Mouse macrophagesTransfection with curcuminRat AD model↑ Neuron survival;
↓ Tau phosphorylation
[118]
Mouse macrophagesEV loading with catalase: sonication, extrusion, or saponinMouse PD model↓ Oxidative stress
↑ Neuron survival
[106]
Human ESCsEV loading with paclitaxel: diffusionOrthotopic mouse xenografts↑ Accumulation at the glioma spot
↑ Mouse survival
[147]
Mouse BECsEV loading with paclitaxel or doxorubicin: diffusionXenotransplanted brain cancer zebrafish model↑ Brain cancer cell elimination[148]
Mouse blood serumEV loading with dopamine: diffusionMouse PD model↑ Dopaminergic neurogenesis
↑ Symptomatic performance
[150,151]
PD—Parkinson’s disease; AD—Alzheimer’s disease; ESCs—embryonic stem cells; BECs—brain endothelial cells. ↑ Increase, ↓ Decrease.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ahmed, W.; Kuniyan, M.S.; Jawed, A.M.; Chen, L. Engineered Extracellular Vesicles for Drug Delivery in Therapy of Stroke. Pharmaceutics 2023, 15, 2173. https://doi.org/10.3390/pharmaceutics15092173

AMA Style

Ahmed W, Kuniyan MS, Jawed AM, Chen L. Engineered Extracellular Vesicles for Drug Delivery in Therapy of Stroke. Pharmaceutics. 2023; 15(9):2173. https://doi.org/10.3390/pharmaceutics15092173

Chicago/Turabian Style

Ahmed, Waqas, Muhammed Shibil Kuniyan, Aqil Mohammad Jawed, and Lukui Chen. 2023. "Engineered Extracellular Vesicles for Drug Delivery in Therapy of Stroke" Pharmaceutics 15, no. 9: 2173. https://doi.org/10.3390/pharmaceutics15092173

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop