Open access peer-reviewed chapter - ONLINE FIRST

Extracellular Vesicles and Diabetic Retinopathy: Nano-Sized Vesicles with Mega-Sized Hopes

Written By

Samar Habib, Alaa M. Mansour, Babak Baban and Khaled Elmasry

Submitted: 17 February 2024 Reviewed: 23 April 2024 Published: 24 June 2024

DOI: 10.5772/intechopen.1005791

Diabetic Retinopathy - Advancement in Understanding the Pathophysiology and Management Strategies IntechOpen
Diabetic Retinopathy - Advancement in Understanding the Pathophys... Edited by Mohd Nawaz

From the Edited Volume

Diabetic Retinopathy - Advancement in Understanding the Pathophysiology and Management Strategies [Working Title]

Dr. Mohd Imtiaz Nawaz

Chapter metrics overview

15 Chapter Downloads

View Full Metrics

Abstract

Diabetic retinopathy (DR) is a major health problem, being the leading cause of vision loss in the working-age group. Novel discoveries facilitating the early diagnosis and curative treatment of DR are still greatly needed. Extracellular vesicles (EVs) are nano-sized vesicles released from different kinds of body cells including various retinal cells for intercellular communication. EVs recently attracted attention because of their non-immunogenic, biocompatible, and biodegradable characteristics. Recent studies investigated EV cargoes in DR patient samples, animal models, and in vitro models of DR. Although a thorough understanding of the trafficking of EVs in the retina still needs extensive investigations, these studies will pave the way for the discovery of novel biomarkers for early diagnosis of DR. Furthermore, EVs-based therapeutics could be of great interest in the treatment of DR. They are presented as promising delivery tools for both pharmacological and genetic therapies. Here, we highlighted the studies that discussed the role of EVs in both inflammation and angiogenesis as two main pathophysiological events occurring in DR. We also tried to portray what we know so far about the role of EVs in DR as a diagnostic, prognostic, and therapeutic tool.

Keywords

  • diabetic retinopathy
  • extracellular vesicles
  • exosomes
  • microvesicles
  • inflammation
  • angiogenesis
  • retina

1. Introduction

Diabetic retinopathy (DR) is a diabetes-related complication characterized by various molecular, metabolic, functional, and structural changes in the retina. It is considered the leading cause of acquired preventable sight loss in the working-age population worldwide. It causes visual impairment that negatively impacts the individual’s quality of life [1, 2, 3, 4, 5]. DR has several risk factors such as the duration of diabetes mellitus (DM), poor glycemic control, hypertension, hyperlipidemia, obesity, smoking, alcohol intake, and pregnancy [6, 7, 8]. Moreover, genetic determinants have a role in the development and progression of DR [9].

Regular DR screening is recommended for diabetic patients to recognize high-risk DR characteristics. Therefore, it is essential to detect cases that require timely full ophthalmic examination and treatment for permanent visual loss prevention and to reduce the new cases of blindness. Additionally, screening is useful for identifying cases at the risk of cardiovascular disease, which expands their role beyond the prevention of sight-threatening disease. Screening is done using diagnostic modalities for DR which are important for the detection and classification of DR. Current diagnostic modalities include ophthalmoscopy and retinal photography, standard color fundus photography, multicolor fundus photography, fundus fluorescein angiography, optical coherence tomography, optical coherence tomography angiography, ultrawide-field imaging systems, swept-source wide-field optical coherence tomography angiography, smartphone-based imaging, and artificial intelligence systems [10].

There are several stages for DR. The reduction in visual acuity (VA), distortion of visual images, and sight loss can occur at any stage of the disease. In the early stages, the disease may remain asymptomatic, but later on, retinal examination reveals progressive retinal damage. This retinal damage appears firstly as microaneurysms, followed by the appearance of cotton wool spots, retinal hemorrhages, and intraretinal microvascular abnormalities. Eventually, this may lead to neovascularization with fragile new blood vessels, so they can be easily damaged, causing vision obstruction. Neovascularization can also detach the retina, resulting in blindness [4].

Regarding the pathophysiology of DR, oxidative stress and inflammation occupy a major role in the development and progression of DR. Sustained high-circulating glucose level activates several inflammatory pathways such as the protein kinase C pathway, the hexosamine biosynthetic pathway, and advanced glycation end-products accumulation (Figure 1). This chronic hyperglycemia increases reactive oxygen species (ROS) exacerbating retinal oxidative stress and inflammation [2, 11]. This leads to a vicious circle of ROS inducing an increase of the inflammatory factors, and then, inflammation induces ROS generation. The nuclear factor κappa-B (NF-κB) is activated also in DM. It is a transcription factor regulating many cytokines, for example, interleukin (IL)-1β, IL-8, tumor necrosis factor (TNF)-α, and several chemokines [4, 12, 13]. Moreover, oxidative stress and inflammation can cause epigenetic modifications. These epigenetic modifications are rapid and transmissible to the next generation [4, 14].

Figure 1.

Schematic diagram showing different pathways involved in DR pathogenesis. These pathways induce oxidative stress and inflammation that lead to diabetic retinopathy. NADPH; Polyol pathway downregulates the nicotinamide adenine dinucleotide phosphate, AGEs; advanced glycation end products, NF-κB; nuclear factor κappa-B, IL; interleukin, TNF- α; tumor necrosis factor-α, GAPDH; glyceraldehyde 3-phosphate dehydrogenase.

The upregulation of inflammatory mediators in DM, together with growth factors such as vascular endothelial growth factor (VEGF), cause blood-retinal barrier (BRB) breakdown, vascular damage, and neuro-inflammation, as well as pathological neovascularization (NV) noticed in DR [15, 16]. VEGF is a major pathogenic factor in DR. Upregulated VEGF induces the expression of more inflammatory cytokines, inducing endothelial dysfunction and breakdown of the BRB. The sub- and intraretinal fluid accumulation causes macular thickening, known as diabetic macular edema (DME), and is considered a complication of DR [2, 17]. As DR is a neurovascular complication of diabetes, non-vascular cells, including retinal neurons and glial cells, are also affected, showing many abnormalities or accelerated apoptosis. Retinal microglia and astrocytes are two main sources of ROS causing oxidative stress in chronic neurodegeneration [4, 18, 19]. Diabetic retinal neurodegeneration is shown as a significant thinning of the inner retinal layers, including the retinal nerve fiber layer, and ganglion cell layer [20, 21, 22].

The current therapies available for DR aim to stop the inflammatory response, prevent retinal NV, and stabilize the BRB [23]. There is a wide range of therapies for DR which are always under development. The selection of the treatment modality depends on several factors. For instance, vitrectomy, where the blood and scar tissue is removed along with vitreous removal, is important in the management of certain cases in DR, for example, non-clearing vitreous hemorrhages. Laser photocoagulation of the abnormal leaky blood vessels is also used to treat pathological NV [24].

Pharmacotherapy has also been used and it produces more progress in the treatment of DR than laser or surgical therapy, but its cost is still a challenge [24]. Corticosteroids, such as dexamethasone, triamcinolone, and fluocinolone, are potent anti-inflammatory agents. They antagonize various pathologic inflammatory mediators such as TNF-α, IL-1β, and IL-6 [25]. As VEGF plays a major role in the pathogenesis of DR, it is considered a critical therapeutic target. Intravitreal injection of anti-VEGF therapy such as bevacizumab, brolucizumab, ranibizumab, and aflibercept can improve non-proliferative DR (NPDR) and prevent the onset of angiogenesis and DME [20, 26, 27, 28, 29]. Unfortunately, not all patients respond properly to anti-VEGF such as in the case of DME with subretinal fluid [2]. Another treatment modality, depending on the integration of laser therapy with pharmacotherapy, may provide a more durable and effective response [24]. It can also result in optimum clinical efficacy with reduced treatment burden for the patient. In proliferative DR (PDR), the combination treatment between anti-VEGF and pan-retinal photocoagulation may be superior to monotherapy [30].

As DR is a polygenetic disease influenced by both environmental and genetic factors, gene therapy is considered a potential alternative therapy for DR. Generally, gene therapy is proposed to introduce genetic material into the cells to compensate for defective genes or deliver therapeutic transgenes. It has several benefits, such as prolonged effect with less frequency of intravitreal injection [31, 32]. One of the methods used for gene delivery is extracellular vesicles (EVs), as it is considered an immune-friendly gene therapy-delivering approach.

Generally, EVs are lipid-bound structures released by cells into the extracellular space carrying bioactive components that interact with target cells, causing subsequent modification. They can be detected in different biological fluids. EVs represent important tools of intercellular communication that allow the exchange of various messages including proteins, lipids, and nucleic acids between adjacent and/or distant cells. These intercellular messages have critical rules in normal homeostasis and disease development [33]. Therefore, EVs are considered novel diagnostic/prognostic biomarkers, promising drug delivery vehicles, and unique tools for targeted therapy [34, 35]. EVs can effectively deliver their cargo, including drugs or nucleic acids, to targeted tissues or organs as they possess several natural advantages, such as stability in circulation, transportation capability, and natural barriers-traversing capacity [36].

According to the pathways involved in their biogenesis and release, size, contents, and functions, EVs are classified into exosomes, microvesicles (MVs), and apoptotic bodies [37]. Exosomes are generated via the inward budding of the limiting membrane of early endosomes during maturation into multivesicular bodies (MVBs). They vary in size from 50 to 150 nm [38]. Their formation and release are regulated by endosomal sorting complexes required for transport (ESCRT) [39]. Nonetheless, ESCRT-independent pathways have also been identified, where sphingomyelinase converts sphingomyelin into ceramides, which allows the sorting of the cargoes in the exosomes and enhances the formation of curvatures in the membrane [40]. Exosomes were shown to have higher levels of transmembrane proteins and glycoproteins compared to whole-cell lysate [41]. Based on the cholesterol contents, MVBs are degraded by lysosomes, or they merge with the plasma membrane and liberate their exosome contents into the extracellular space [42, 43]. Exosomes interact with the target cells by micropinocytosis, endocytosis, or they fuse with the cell membrane and deliver their contents into the cell [44]. Figure 2 describes the biogenesis and uptake of exosomes.

Figure 2.

A schematic diagram illustrating different steps of EVs biogenesis and release from donor cells. EVs uptake by the recipient cells can occur via endocytosis, pinocytosis, or membrane fusion.

Microvesicles are formed by external budding and then scission from the plasma membrane of both healthy and damaged cells. Their size ranges between 50 and 500 nm, and up to 10 μm in cases of cancer-derived EVs, known as oncosomes. Cytoskeletal components, molecular motors, and fusion machinery were shown to contribute to MVs formation [45]. In comparison to exosomes, MVs tend to contain higher levels of proteins with post-translational modifications [41]. Different mechanisms are adopted to mediate MVs-cellular communications such as receptor-ligand interaction, fusion of MVs membrane with that of the recipient cell, endocytic-mediated internalization of MVs contents, and membrane protease cleavage resulting in the release of MVs soluble proteins, which interact with specific receptors on the target cells [46].

Regarding apoptotic bodies, they are typically larger, ranging from 50 to 5000 nm. They are generated during the process of programmed cell death (apoptosis) as blebbing from the plasma membrane of cells [47]. They enclose intact organelles, chromatin, and proteins [48]. In contrast to exosomes and MVs, the proteomic analysis of apoptotic bodies is similar to that of cell lysate [49]. The features of different subtypes of EVs were summarized by Hu et al. [50].

The basics of uptake and intercellular trafficking of different subtypes of EVs are thought to be shared despite different sizes and components [36]. Inside the recipient cell, endocytosed EVs either follow the degradation pathway or release their contents into the cytoplasm [35]. Van der Pol et al. [51] and Lv et al. [52] have reviewed the functions of exosomes and MVs and highlighted their crucial roles in cell-to-cell communication and signaling, particularly in the immune response, tumorigenesis, thrombus formation, and waste removal. Figure 3 portrays retinal intercellular communication via EVs [51, 52].

Figure 3.

Different retinal cells use EVs as a tool for intercellular communication. This diagram illustrates different sources of color-coded EVs from various retinal cells.

In this book chapter, we briefly discuss the EVs’ isolation. We emphasize EVs originating from various sources, either intraocular or extraocular sources, and explain how they contribute to inflammation and angiogenesis, as two major arms of DR pathophysiology. Additionally, we highlight the EVs as promising diagnostic biomarkers and therapeutic tools in the field of DR. Finally, we list the related challenges and future recommendations.

Advertisement

2. Isolation and characterization of EVs

As most isolation methods depend on the distinction of EVs according to the size, and due to size overlap between exosomes and MVs, it is hard to obtain a pure population of EVs subtypes [53]. Commonly used isolation methods include immunoprecipitation, polymer-based precipitation, ultrafiltration, ultracentrifugation, density gradient, microfluidics, and size exclusion chromatography. An interesting study by Helwa et al. [54] compared ultracentrifugation versus commercially available kits for exosome isolation and showed that even with limited volumes of biological samples, commercially available isolation kits were feasible alternatives to the ultracentrifugation technique. The principle of the techniques, advantages, disadvantages, yield, and purity are neatly tabled by Massoumi et al. [55]. Regarding identification and characterization, tetraspanins, such as CD9, CD63, and CD81, are generally used as markers for exosomes. CD40, selectins, integrins, and annexin 1 are used for MVs characterization. As far as apoptotic bodies, DNA, histone, and phosphatidylserine are commonly used [55, 56, 57]. Electron microscopy (EM) studies including transmission EM, scanning EM, or immune-gold labeling using different EVs markers are generally used as an additional confirmatory step for EVs characterization [58, 59]. Nanoparticle tracking analysis is commonly used to report the concentration and the size range of EVs in isolated samples [54].

Advertisement

3. Role of EVs in inflammation

Inflammation and immune dysfunction result from chronic hyperglycemia and hyperlipidemia, are believed to be key mechanisms in the development of DR. Increased levels of inflammatory cytokines and angiogenic factors were noticed in plasma EVs from diabetic patients, which find their way to the retina because of increased permeability of BRB and are known to contribute to the pathophysiology of DR [60]. Our previous work highlighted the pathway analysis of retinal pigment epithelium (RPE)-derived exosomal microRNAs (miRNAs or miRs) that showed their involvement in different cellular pathways related to the development of DR such as oxidative stress, endoplasmic reticulum (ER) stress, and VEGF signaling [61]. Our ongoing research noticed a differential exosomal miRNA expression profile of retinal endothelial cells treated with proinflammatory lipid mediators. Inflammatory mediators such as TNF-α, thrombin, and C-reactive protein (CRP) were found to enhance endothelial cells (ECs) to form MVs in vitro. These MVs promote inflammation by triggering target cells to produce ROS, leading to oxidative stress [62, 63, 64]. High glucose causes increased NADPH oxidase activity in endothelium-derived microparticles, contributing to disturbed retinal barrier functions [65]. EVs derived from RPE under oxidative stress promoted apoptosis and inflammation through Apaf1/caspase-9 axis [66]. Additionally, Tokarz et al. [67] found a correlation between chemokine receptor (CCR)-5 positive MVs with NPDR progression.

Platelet-derived MVs from patients with DR enhanced the interaction between monocytes and endothelial cells [68]. Also, in diabetic patients, high levels of CXCL-10 in platelet-derived EVs stimulated the Toll-like receptor (TLR)-4 pathway, triggered ROS production, inhibited superoxide dismutase (SOD) activity, and lessened tight junction proteins, with the net result of increased BRB permeability [69]. Additionally, exosomes derived from platelet-rich plasma from diabetic rats enhanced the proliferation and fibrogenic activity of human retinal Müller cells (hMCs) through PI3K/AKT signaling [70].

Microglia are the principal immune cells in the retina which respond to paracrine signals carried by EVs. DM promotes a chronic inflammatory status in the body, which causes polarization of the microglia to the proinflammatory M1 phenotype [71]. Furthermore, M1-polarized microglia accumulate around the capillaries and release EVs, which heighten neuro-inflammation and increase vascular permeability [72, 73]. Beltramo et al. [74] reported a significant increase in inflammatory markers, including chemokine ligand (CCL)-2, matrix metalloproteinase (MMP)-2, and MMP-9, in M1-polarized microglia-derived EVs. A significant increase in the miRNAs related to inflammation and angiogenesis (miR-21 and miR-155) was also marked. Interestingly, exposure of human retinal pericytes (HRPs) and Human retinal endothelial cells (HRECs) to EVs derived from M1-polarized microglia caused a significant increase in their proliferation, apoptosis, migration, and ROS production. In addition, HRPs displayed significant production of IL-1β, IL-6, MMP-9, CCL-2, and VEGF, while HRECs exhibited significant release of TNF-α, angiopoietin (Ang)-2, VEGF, and platelet-derived growth factor (PDGF)-B, when exposed to M1-polarized microglia-derived EVs. Of note, Müller glia cells-derived exosomes were found to aggravate vascular dysfunction under high glucose through exosomal miR-9-3p, which is transferred to retinal endothelial cells and binds to the sphingosine-1-phosphate receptor S1P1 coding sequence, which subsequently activates VEGFR2 phosphorylation [75].

The complement system is a key mechanism deployed by the innate and adaptive immune responses and causes damage to opsonized pathogens and cells via deposition of the membrane attack complex (MAC) on the plasma membrane, resulting in osmotic disturbance and lysis of the target cell [76, 77]. The expression of MAC was marked in the chorio-capillaries and around larger choroidal vessels in patients with DR [78]. This complement-mediated damage is attributed to continuous complement activation, in addition to the lack of complement regulatory proteins on the retinal endothelium [79]. Interestingly, Huang et al. [80, 81] reported robust complement activation in the plasma of diabetic rats by immunoglobulin (Ig) G-enriched EVs. Further, these IgG-laden EVs caused MAC deposition and lysis of HRECs in vitro, which was abrogated by the depletion of EVs from the plasma.

Advertisement

4. Role of EVs in angiogenesis

The involvement of EVs in the pathogenesis of DR, particularly in establishing microvascular dysfunction and angiogenesis, has been investigated in numerous studies. Although RPE-derived EVs are believed to induce protective effects in physiological states and during the early stages of DR, EVs originated from stressed ARPE-19 cells enhanced angiogenesis, through upregulation of VEGF receptors on HUVEC cells [82]. A longitudinal cohort study conducted by Wu et al. [83] reported the significant link between EVs levels of VEGF-A, the diabetes situation, and parameters of insulin resistance, in diabetic patients when compared to euglycemic controls. Moreover, when endothelial cells were treated with EVs from diabetic patients, the formation of lamellipodia increased and led to enhanced migration of endothelial cells.

Plasma-derived EVs from patients with DR were capable of inducing retinopathy indices in retinal microvascular in vitro models in the form of detachment and migration of pericytes, increased permeability, and new vessel-like structure formation [84]. TNF-α-induced protein 8 (TNFAIP8) was demonstrated in the small EVs (sEVs) isolated from DR patients’ plasma and was linked to increased viability, proliferation, migration, and tube formation of HRECs [85].

Circulating EVs were reported to increase in diabetic patients and to induce phenotypic changes similar to those of DR in different study models owing to their bioactive cargoes, especially miRNAs [86]. Free miRNAs in the plasma are liable to degradation by enzymes; however, EVs’ lipid membrane protects them [74]. Plasma-derived EVs from patients with DR did not display differences in the surface markers when compared to diabetic patients with no DR or healthy controls; however, microarray analysis revealed significant changes in several miRNAs [84]. One of the important miRNAs reported to promote angiogenesis is miR-30b, which was found to be prominently enriched in plasma-derived EVs and was the most differentially expressed miRNA in the retinal tissues of mice with PDR, compared to control mice. miR-30b exerts its pro-angiogenic effect on retinal microvascular endothelial cells (RMECs) through repression of sirtuin (SIRT)-1; the latter was stated to preserve vascular integrity of the retina and protect against the development of DR in the early phases [87, 88]. Besides, upregulation of miR-30a-5p was recognized in the vitreous specimens of patients diagnosed with PDR [89]. It promotes angiogenesis by enhancing endothelial cells’ motility and tube formation [90]. miR-21 was also found to enhance angiogenesis through repression of peroxisome proliferator-activated receptor-α (PPARα) [91]. In addition, the expression of miR-30b-5p, miR-21-3p, and hypoxia-inducible factor (HIF)-1α increased, and that of miR-150-5p decreased in EVs derived from DR patients, compared to healthy controls and diabetic patients with no DR [92]. Noteworthy, miR-150 was stated to inhibit the migration of endothelial cells (ECs) through inhibition of numerous angiogenic factors [93].

Pericytes regulate the blood capillaries tone, control the interchange of molecules between the blood and the neuroretina, and limit the proliferation and motility of ECs; thus, their loss leads to the development of angiogenesis, which is a key mechanism in the early stages of DR [94]. EVs control the interplay between pericytes and ECs in DR, where circular RNA (circRNA) cPWWP2A is shuttled from pericytes to ECs by EVs, inducing retinal vascular malformations through enhanced expression of Ang-1, occludin, and SIRT-1 [95]; however, a protective role of Ang-1 in DR has been described [96]. EVs derived from microglia and photoreceptors were reported to increase in the vitreous of PDR patients and were found to promote ECs proliferation and angiogenesis [97]. Maisto et al. [98, 99] demonstrated a decrease in the anti-angiogenic miRNAs (miR-20a-3p, miR-20a-5p, miR-20b, and miR-106a-5p), together with increased expression of VEGF in retinal photoreceptors/ARPE-19 cells and their derived EVs, when challenged with high glucose.

The role of MVs in the development of DR was summarized by Zhang et al. [46], where the diabetic milieu causes MVs of platelet origin to activate MMPs, which cause mitochondrial damage and release of cytochrome c, which in turn enhances apoptosis of capillary endothelial cells and consequently, capillary hypoxia, which leads to NV over time. MVs of monocyte origin were claimed to cause damage to RPE, compromising the barrier function and increasing the permeability. Noteworthy, monocyte-derived MVs were the most prominent during the transition of DR from the non-proliferative to the proliferative stage, where abnormal leaky blood vessels are formed [100].

Significantly, mesenchymal stem cells (MSCs) were established to contribute to the homeostasis and the pathology of different tissues, including the retina, through the paracrine effect of EVs [101]. Beltramo et al. [102] reported that HRPs display enhanced detachment when exposed to EVs isolated from MSCs, which were grown in high glucose and/or hypoxia to mimic diabetic conditions, regardless of the origin of MSCs. The detached cells were viable and were able to re-attach again, suggesting that these cells tend to localize in the walls of the newly formed vessels for stabilization. They also described increased permeability of the blood barrier, and enhanced formation of vessel-like structures in HRP/EC co-cultures on Matrigels when treated with the same EVs. Moreover, they attributed the effect of MSCs-derived EVs on HRPs and ECs to the active form of MMP-2, which causes degradation of the proteins in the extracellular matrix, facilitating pericyte loss and angiogenesis. Interestingly, pancreatic β cells were proved to release EVs rich in miR-15a, which spread to distant sites and promote DR through induction of oxidative stress and apoptosis in Müller cells [103].

It is worth mentioning that procoagulant MVs in patients with DR correlate with the duration of diabetes [104]. Phosphatidyl serine (PS)-positive MVs are linked to enhanced coagulant activity in patients with PDR and NPDR [105]. Additionally, endothelial-derived MVs are considered a risk factor for the development of thrombo-embolic DR [106]. The roles of EVs in the initiation and progression of DR are summarized in Table 1.

Source of EVsMethod of isolation and/or purificationRole of EVs in DRRefs.
Extraocular cell-derived EVsPlasmaIsolated by ultracentrifugation and purified by OptiPrep Density GradientCirculating IgG-laden exosomes are responsible for the activation of the complement cascade that subsequently upregulates proinflammatory cytokines and chemokines and causes MAC deposition and lysis of HRECs in vitro.[80, 81]
Isolated using total exosome isolation reagent for cell culture mediaPlasma-EVs transferred miR-30b to retinal microvascular endothelial cells (RMECs), where miR-30b increases VEGF expression by downregulating SIRT1. This enhances angiogenesis and proliferative DR.[87]
Platelet-rich plasma (PRP)Isolated by ultracentrifugationHyperglycemia elevates the level of circulating PRP-Exos which activates the TLR4 pathway through the release of CXCL10. This led to endothelial injury and dysfunction in early DR.[69]
Isolated by ultracentrifugationPlatelet rich plasma derived exosomes in hyperglycemia enhance both the proliferation and fibrogenic activity of Müller cells through PI3K/AKT signaling.[70]
Pancreatic β cellsIsolated using total exosome isolation reagent for cell culture mediaExposing Müller cells to exosomes derived from pancreatic β-cells under high-glucose conditions caused miR-15a overexpression in Müller cells and increased the oxidative stress and apoptotic cell death.[103]
Intraocular cell-derived EVsRetinal pigment epithelial (RPE) cellsIsolated by multi‐step differential centrifugationExosomes derived from RPE cells under oxidative stress increased Apaf1 expression, so oxidative injury and cell apoptosis increased.[66]
Isolated using total exosome isolation reagent for cell culture mediaRPE cells cultured in a high glucose level led to ROS formation and release of VEGF containing exosomes.[99]
Retinal Astrocytes (RACs)Isolated by ultra centrifugationApoptosis and autophagy levels in RACs were positively correlated with the oxidative stress level, and the exosomes generated from RACs under oxidative stress condition. These exosomes can promote endothelial cell proliferation, migration, and tube formation.[57]
Retinal PhotoreceptorIsolated using total exosomes isolation reagentHigh glucose increased VEGF levels and decreased anti-angiogenic miR-20a-3p, miR-20a-5p, miR-106a-5p, and miR-20b expression in retinal photoreceptor-derived EVs due to ROS-induced NF-κB signaling.[98]
Müller glia cellsIsolated using ultracentrifugationMüller glia-derived exosomes aggravate vascular dysfunction under high glucose, through exosomal miRNA-9-3p which is transferred to retinal endothelial cells and binds to the sphingosine-1-phosphate receptor S1P1 coding sequence, with subsequent activation of VEGFR2 phosphorylation.[75]
Retinal PericytesNot mentionedPericytes transferred cPWWP2A, miR-579, and angiopoietin 1/occludin/SIRT1 to endothelial cells by exosomes. These transferred materials induced retinal vascular dysfunction.[95]

Table 1.

Role of EVs in the initiation and progression of the pathogenesis of DR.

Advertisement

5. How can EVs help in the diagnosis of DR?

The clinical diagnosis of DR is challenging since the disease is asymptomatic during the early stages. Development of symptoms in the form of blurred vision may occur with more progressive pathology [86]. Given that EVs reflect the molecular profile of the parent cells and the important role played by EVs, particularly the miRNAs shuttled by EVs in the pathophysiology of DR [107], it appeared valuable to use them as diagnostic biomarkers. Several studies aimed at the detection of EVs-miRNAs exclusive to DR.

As mentioned before, cPWWP2A circRNA is shuttled by EVs derived from pericytes in DR, and it is thought to have a protective role in refining pericytes-endothelial cells communication and amelioration of vascular pathology. This circRNA can be used as a diagnostic biomarker [95]. Likewise, EVs positive for miR-15a [103], IgG [108], or TNFAIP8 [85], all can be used for the diagnosis of DR. EVs with increased expression of miR-30b-5p, miR-21-3p, miR-26b-5p, or decreased expression of miR-150-5p were identified in the sera of DR patients [92, 109]. Additionally, EVs carrying CCR-5 can be used to detect NPDR progression [67]. Remarkably, Mighty et al. [110] confirmed the expression of junction plakoglobin protein (JUP) in the EVs isolated from retinal tissue explants and urine of DR patients but not in healthy controls. This was the first study to provide a novel non-invasive diagnosis for DR through urinary specimens. Table 2 shows examples of EV-derived cargoes that could be used as biomarkers for DR.

EVs markerOriginRoleReference
cPWWP2APericytesRefining pericytes-endothelial cells communication and amelioration of vascular pathology[95]
miR-15aPancreatic β cellsInduction of oxidative stress and apoptosis in Müller cells[103]
IgGPlasma of diabetic ratsRobust complement activation MAC deposition and lysis of HRECs in vitro[80]
TNFAIP8Plasma of DR patientsIncreased viability, proliferation, migration, and tube formation of HRECs[85]
CCR-5Plasma of DR patientsRegulates trafficking and effector functions of immune cells NPDR progression[67]
JUPRetinal tissue explants and urine of DR patientsEarly screening biomarker for DR diagnosis[110]

Table 2.

Examples of EVs cargoes that could serve as potential biomarkers for DR.

Interestingly, some studies have proved the effectiveness of certain miRNAs as diagnostic markers; however, these studies did not consider their expression in EVs. For instance, miR-181c, miR-21, and miR-1179 were reported to increase significantly in the sera of PDR patients when compared to NPDR patients [111]. Detection of such miRNAs in serum EVs may provide higher diagnostic values. Similarly, Friedrich et al. [112] found higher levels of six miRNAs in the vitreous of PDR patients when compared to non-diabetic patients namely, hsa-miR-23b-3p, hsa-miR-20a-5p, hsa-miR-142-3p, hsa-miR-185-5p, hsa-miR-362-5p, and has-miR-326. The first one, hsa-miR-23b-3p, was found to be downregulated in anti-VEGF-treated patients in comparison to non-treated PDR patients. On the other hand, miR-126 decreased in DR patients and could serve as a diagnostic biomarker for PDR [113]. We recommend conducting future studies to verify the expression of these miRNAs in circulating EVs, which might spot the time of alteration in their levels and allow their use as diagnostic and prognostic markers through non-invasive sampling procedures.

Advertisement

6. Are EVs a promising new therapeutic tool for DR?

Impairment and loss of vision are unavoidable if DR is left untreated. During the initial stages, proper control of blood glucose levels may prevent the deterioration of the disease. Treatment of hypertension and hyperlipidemia is also important; however, medical intervention is necessary in the late stages. Anti-VEGF, corticosteroids, lipid-lowering drugs, argon laser photocoagulation, and vitrectomy are commonly used, as reviewed by Martins et al. [86]. Immunotherapy such as anti-TNF-α and anti-IL-1β are being tested [114, 115]. Although current interventions may help prevent or delay the sequelae of the disease, lack of curative treatment, adverse reactions, and ineffectiveness of these regimens in some patients are still considerable challenges [116]. Therefore, it is important to seek novel and more tolerable therapies, such as EVs, which can deliver many factors including miRNAs and medications to the retina, in addition to enhancing the efficacy of certain drugs (Figure 4) [117].

Figure 4.

EVs derived from intra- or extra-ocular sources can be used as a therapy for DR directly, as a drug delivery nanocarrier, or engineered EVs with elevated levels of targeted miRNAs.

Intriguingly, intraocular injection of exosomes derived from rabbit adipose tissue MSCs in diabetic rabbits resulted in well-organized retinal layers. The link between increased miR-222 in MSCs-derived exosomes and the regenerative events in the retina was highlighted [118]. In addition, Zhang and colleagues [119], used umbilical cord MSCs-derived exosomes as a vehicle for miR-126. Intravitreal injection of these exosomes in diabetic rats suppressed retinal inflammation. The authors confirmed the role of miR-126-over expression in MSCs-derived exosomes in the reduction of high-mobility group box (HMGB)-1 and inflammasome, which resulted in amelioration of retinal inflammation. Gu et al. [120] reported that MSCs-derived EVs, which shuttle miR-192, ameliorated retinal inflammation and angiogenesis. Reddy et al. [121] reported that intravitreal injection of MSCs-derived sEVs loaded with anti-VEGF caused prolonged inhibition of VEGF and leukostasis and decreased exudates in diabetic rats for more than 2 months following injection, supporting the value of sEVs as drug delivery system. Furthermore, the anti-oxidant and anti-apoptotic effect of MSCs-derived EVs was reported in an animal model of DR. The in vitro experiments attributed this therapeutic effect to neuronal precursor cell-expressed developmentally downregulated 4 (NEDD4), which regulates PTEN/AKT/NRF2 pathway [122]. On the other hand, EVs derived from MSCs, which were grown in high glucose, were claimed to accelerate vascular dysfunction [102]; therefore, the use of MSCs-derived EVs in the treatment of DR needs more extensive studies.

Interestingly, exosomes derived from retinal astroglial cells (RACs), but not from RPE, inhibited vessel leakage and choroidal neovascularization (CNV) in a laser-induced CNV model. These anti-angiogenic properties may be useful when applied in DR models [123]. RPE-derived exosomes were found to inhibit endothelial-to-mesenchymal transition (EMT) and retinal fibrosis due to their miR-202-5p content, which inhibits transforming growth factor/Smad signaling [124]. Of note, our study showed that RPE-treated with homocysteine, which is reported as a risk factor for retinopathies, released exosomes with micro-RNA cargo close to the miRs profile noticed in the diabetic mouse retina, such as miR-17, let-7, and miR-122 [61]. Recently, Huang et al. [125] analyzed sEVs derived from human embryonic stem cell-derived retinal organoids (hERO-RPCs) and found them to delay the degeneration of photoreceptors, inhibit gliosis of Müller glia cells, and support retinal functions in RCS rats. They attributed these effects to miRNA-mediated downregulation of nuclear factor I transcription factors B (NFIB).

Lymphocyte-derived microparticles (LMPs) were found to contain high levels of miR-181a. This miRNA interferes with VEGF signaling in HREC; therefore, it could be used for the treatment of DR [126]. Besides, control of complement activation was achievable through the exclusion of IgG-laden plasma EVs, which led to the protection of the ECs from cytolytic damage [80]. A recent study by Lamb et al. [127] investigated the Müller glia cells-derived EVs and highlighted their neuroprotective functions through enrichment with miRNAs known to support the stem cells, such as miR-21 and miR-16, and miRNAs which keep the retinal homeostasis; such as miR-125b, miR-9, and let-7 family. Mathew et al. [128] treated retinal cells suffering from ischemic insult with functionally engineered EVs overexpressing miR-424 (FEE424), and found them to achieve anti-inflammatory and neuroprotective functions. They also used FEE424 in an animal model of retinal ischemia and reported that retinal functions were restored.

Other miRNAs were confirmed to have a protective effect on the retinal tissue. For instance, Shaker et al. [129] found that miR-20b and miR-17-3p decrease significantly in the sera of both PDR and NPDR patients, in comparison to healthy controls. miR-20b and miR-17-3p are believed to regulate the expression of HIF-1α and VEGF-A genes. Another study reported downregulation of four miRNAs in the sera of DR patients namely, miR-338-3p, miR-4448, miR-9-5p, and miR-485-5p [130]. These miRNAs regulate inflammation, oxidative stress, and angiogenesis through the regulation of dozens of genes. miR-200b expression in different DR animal models revealed controversial results; nevertheless, McArthur et al. [131] reported its decrease in the retinas and endothelial cells of diabetic rats and linked this decrease to enhanced VEGF expression. Blum et al. [132] confirmed the link between decreased levels of miRNA-423 and increased levels of VEGF in the sera of DR patients. Also, Qin et al. [113] highlighted miR-126 lower levels in DR patients. While EVs were not included in these studies, it is possible to establish a drug delivery system using those EVs to deliver such deficient miRNAs.

In contrast, some studies reported higher values of certain miRNAs in DR patients without the inclusion of EVs in these studies. For example, miR-181c, miR-21, and miR-1179 were proven to increase the sera of DR patients and were known to play important roles in ischemia and angiogenesis [111]. Using EVs as vehicles to deliver such deficient miRNAs may be a valuable drug delivery tool in DR. Sun et al. [133] developed engineered MSCs-EVs with increased levels of miR-5068 and miR-10228 as therapeutic molecules. They used both MSCs-EVs and engineered ones to treat two diabetic animal models, including db/db mice and STZ-induced diabetic rats. The presence of miR-5068 and miR-10228 in both EVs targeted the HIF-1α/EZH2/PGC-1α pathway. However, the engineered MSCs-EVs showed more enhanced retinal repair efficiency than MSCs-EVs.

Advertisement

7. Challenges and future recommendations

The therapeutic potential of EVs requires the construction of clinical-grade EVs, where enormous quantities, in addition to preserved phenotypes, are necessary. As different cell culture procedures aim at increasing the harvest, such as using different stimuli to expand the production or using 3D culture technology, the phenotype of the cultured cells and, consequently, their EVs may change [134]. Passage of the cultured cells, media composition, and oxygen levels could affect the cells/EVs number and quality [135]. The use of the third to fifth passage was recommended to preserve consistency in MSCs [136]. Regarding media composition, while fetal bovine serum was found to influence the behavior of cells, serum-free media changed the protein content of EVs [137]. Platelet lysate with an EV-depleted medium was reported to preserve the phenotype of MSCs and the RNA profile of their EVs [138].

Another challenge is the isolation of EVs. Although several techniques are available, there is no standardized technique for isolation of clinical-grade EVs. Low yield in ultracentrifugation, low purity, or chemical contamination in the other techniques interfere with their use in such settings [50]. Ultrafiltration followed by size exclusion chromatography was confirmed to obtain a higher yield of EVs and preserve their phenotype [139, 140]. Cryopreservation using glycerol and dimethyl sulfoxide (DMSO) was found to disintegrate EVs [141]. Using phosphate-buffered saline (PBS) was reported to form calcium nanoparticles, which intervene with quantification of EVs [142]. Concerning the temperature of storage, different studies reported different results, such as −20 or −80° C [141, 143]. To extend the shelf life and decrease the cost, lyophilization was proposed, with a storage temperature of 4°C [144].

Directing EVs to specific cells is another challenge. While the factors that control the targeting of EVs to certain cells are not fully described, surface molecules, the physiological status of the target cells, and the route of administration play important roles in the process [50, 118]. Using certain labels to track EVs was confirmed to enhance their targeting of certain cells [145, 146]. Improving the therapeutic properties of EVs to reach the target cells is required to decrease the dose or the frequency of administration. Priming, loading, engineering, artificial EVs, and bio nanotechnology were developed for that purpose [147].

Finally, the safety of EV-based therapy is crucial. Although cell therapy may induce tumorigenesis, EVs have been shown to be well tolerated in several animal models. Immunogenicity and toxicity monitoring in humans must be extensively studied [53].

Advertisement

8. Conclusion

DR is a neurovascular disease where different retinal cells may be involved in pathophysiological changes occurring throughout the disease course. Retinal endothelial cells exposed to high glucose levels during diabetes may send early warning messages to other retinal cells, initiating various signaling pathways via endothelial-derived EVs. Retinal Müller glia cells, as important master regulators of retinal homeostasis, may talk to other retinal cells via EVs, trying to fight against the high-glucose-induced retinal injury. Other retinal cells may use this salient method of communication to synergize their actions during the disease. Understanding these EVs-carried messages may help in understanding a more detailed picture of the process of DR development. That is why EVs are promising new tools that could carry novel biomarkers for early diagnosis and prognosis of DR. EVs have been used recently as an encouraging drug delivery method in cases of DR. The field of EVs is an emerging area of research that still needs more extensive studies to portray a thorough understanding of EVs’ retinal trafficking, various cargoes, isolation purity, proper characterization, and therapeutic safety.

Advertisement

Acknowledgments

Dr. Khaled Elmasry would like to thank Augusta University, Augusta, GA, USA, for the start-up fund.

Advertisement

Conflict of interest

The authors have no conflict of interest to declare.

References

  1. 1. Frank RN. Diabetic retinopathy. New England Journal of Medicine. 2004;350(1):48-58
  2. 2. Gomulka K, Ruta M. The role of inflammation and therapeutic concepts in diabetic retinopathy-a short review. International Journal of Molecular Sciences. 2023;24(2):1024
  3. 3. Altmann C, Schmidt MHH. The role of microglia in diabetic retinopathy: Inflammation, microvasculature defects and neurodegeneration. International Journal of Molecular Sciences. 2018;19(1):110
  4. 4. Kowluru RA. Cross talks between oxidative stress, inflammation and epigenetics in diabetic retinopathy. Cells. 2023;12(2):300
  5. 5. Ola MS, Nawaz MI, Khan HA, Alhomida AS. Neurodegeneration and neuroprotection in diabetic retinopathy. International Journal of Molecular Sciences. 2013;14(2):2559-2572
  6. 6. Chew EY, Davis MD, Danis RP, Lovato JF, Perdue LH, Greven C, et al. The effects of medical management on the progression of diabetic retinopathy in persons with type 2 diabetes: The action to control cardiovascular risk in diabetes (ACCORD) eye study. Ophthalmology. 2014;121(12):2443-2451
  7. 7. Lachin JM, Genuth S, Nathan DM, Zinman B, Rutledge BN. Effect of glycemic exposure on the risk of microvascular complications in the diabetes control and complications trial--revisited. Diabetes. 2008;57(4):995-1001
  8. 8. Yau JW, Rogers SL, Kawasaki R, Lamoureux EL, Kowalski JW, Bek T, et al. Global prevalence and major risk factors of diabetic retinopathy. Diabetes Care. 2012;35(3):556-564
  9. 9. Arar NH, Freedman BI, Adler SG, Iyengar SK, Chew EY, Davis MD, et al. Heritability of the severity of diabetic retinopathy: The FIND-eye study. Investigative Ophthalmology & Visual Science. 2008;49(9):3839-3845
  10. 10. Vujosevic S, Aldington SJ, Silva P, Hernandez C, Scanlon P, Peto T, et al. Screening for diabetic retinopathy: New perspectives and challenges. The Lancet Diabetes and Endocrinology. 2020;8(4):337-347
  11. 11. Whitehead M, Wickremasinghe S, Osborne A, Van Wijngaarden P, Martin KR. Diabetic retinopathy: A complex pathophysiology requiring novel therapeutic strategies. Expert Opinion on Biological Therapy. 2018;18(12):1257-1270
  12. 12. Yuuki T, Kanda T, Kimura Y, Kotajima N, Ji T, Kobayashi I, et al. Inflammatory cytokines in vitreous fluid and serum of patients with diabetic vitreoretinopathy. Journal of Diabetes and its Complications. 2001;15(5):257-259
  13. 13. Rübsam A, Parikh S, Fort PE. Role of inflammation in diabetic retinopathy. International Journal of Molecular Sciences. 2018;19(4):942
  14. 14. Kowluru RA, Kowluru A, Mishra M, Kumar B. Oxidative stress and epigenetic modifications in the pathogenesis of diabetic retinopathy. Progress in Retinal and Eye Research. 2015;48:40-61
  15. 15. Tang L, Xu GT, Zhang JF. Inflammation in diabetic retinopathy: Possible roles in pathogenesis and potential implications for therapy. Neural Regeneration Research. 2023;18(5):976-982
  16. 16. Wang W, Lo ACY. Diabetic retinopathy: Pathophysiology and treatments. International Journal of Molecular Sciences. 2018;19(6):1816
  17. 17. Tang L, Zhang C, Lu L, Tian H, Liu K, Luo D, et al. Melatonin maintains inner blood-retinal barrier by regulating microglia via inhibition of PI3K/Akt/Stat3/NF-κB signaling pathways in experimental diabetic retinopathy. Frontiers in Immunology. 2022;13:831660
  18. 18. Tan TE, Finkelstein MT, Tan GSW, Tan ACS, Chan CM, Mathur R, et al. Retinal neural dysfunction in diabetes revealed with handheld chromatic pupillometry. Clinical & Experimental Ophthalmology. 2022;50(7):745-756
  19. 19. Kowluru RA, Mishra M. Chapter three—Regulation of matrix metalloproteinase in the pathogenesis of diabetic retinopathy. In: Khalil RA, editor. Progress in Molecular Biology and Translational Science. 148: Academic Press; 2017. pp. 67-85
  20. 20. Tan TE, Wong TY. Diabetic retinopathy: Looking forward to 2030. Frontiers in Endocrinology (Lausanne). 2022;13:1077669
  21. 21. Chen X, Nie C, Gong Y, Zhang Y, Jin X, Wei S, et al. Peripapillary retinal nerve fiber layer changes in preclinical diabetic retinopathy: A meta-analysis. PLoS One. 2015;10(5):e0125919
  22. 22. Sohn EH, van Dijk HW, Jiao C, Kok PH, Jeong W, Demirkaya N, et al. Retinal neurodegeneration may precede microvascular changes characteristic of diabetic retinopathy in diabetes mellitus. Proceedings of the National Academy of Sciences of the United States of America. 2016;113(19):E2655-E2664
  23. 23. Elmasry K, Habib S, Moustafa M, Al-Shabrawey M. Bone morphogenetic proteins and diabetic retinopathy. Biomolecules. 2021;11(4):593
  24. 24. Mansour SE, Browning DJ, Wong K, Flynn HW Jr, Bhavsar AR. The evolving treatment of diabetic retinopathy. Clinical Ophthalmology. 2020;14:653-678
  25. 25. Jeong S, Ku SK, Bae JS. Anti-inflammatory effects of pelargonidin on TGFBIp-induced responses. Canadian Journal of Physiology and Pharmacology. 2017;95(4):372-381
  26. 26. Wells JA, Glassman AR, Ayala AR, Jampol LM, Bressler NM, Bressler SB, et al. Aflibercept, bevacizumab, or ranibizumab for diabetic macular edema: Two-year results from a comparative effectiveness randomized clinical trial. Ophthalmology. 2016;123(6):1351-1359
  27. 27. Brown DM, Wykoff CC, Boyer D, Heier JS, Clark WL, Emanuelli A, et al. Evaluation of intravitreal aflibercept for the treatment of severe nonproliferative diabetic retinopathy: Results from the PANORAMA randomized clinical trial. JAMA Ophthalmology. 2021;139(9):946-955
  28. 28. Wykoff CC, Eichenbaum DA, Roth DB, Hill L, Fung AE, Haskova Z. Ranibizumab induces regression of diabetic retinopathy in Most patients at high risk of progression to proliferative diabetic retinopathy. Ophthalmology Retina. 2018;2(10):997-1009
  29. 29. Maturi RK, Glassman AR, Josic K, Antoszyk AN, Blodi BA, Jampol LM, et al. Effect of intravitreous anti-vascular endothelial growth factor vs sham treatment for prevention of vision-threatening complications of diabetic retinopathy: The protocol W randomized clinical trial. JAMA Ophthalmology. 2021;139(7):701-712
  30. 30. Filho JA, Messias A, Almeida FP, Ribeiro JA, Costa RA, Scott IU, et al. Panretinal photocoagulation (PRP) versus PRP plus intravitreal ranibizumab for high-risk proliferative diabetic retinopathy. Acta Ophthalmologica. 2011;89(7):e567-e572
  31. 31. Raman R, Swathipriya N, Sripriya S. Gene therapy in diabetic retinopathy. In: Genetics of Ocular Diseases. Singapore: Springer; 2022. pp. 187-194
  32. 32. Han J, Lando L, Skowronska-Krawczyk D, Chao DL. Genetics of diabetic retinopathy. Current Diabetes Reports. 2019;19:1-8
  33. 33. Raposo G, Stoorvogel W. Extracellular vesicles: Exosomes, microvesicles, and friends. The Journal of Cell Biology. 2013;200(4):373-383
  34. 34. Bettio V, Mazzucco E, Antona A, Cracas S, Varalda M, Venetucci J, et al. Extracellular vesicles from human plasma for biomarkers discovery: Impact of anticoagulants and isolation techniques. PLoS One. 2023;18(5):e0285440
  35. 35. Liu YJ, Wang C. A review of the regulatory mechanisms of extracellular vesicles-mediated intercellular communication. Cell Communication and Signaling: CCS. 2023;21(1):77
  36. 36. Van Niel G, d'Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nature Reviews Molecular Cell Biology. 2018;19(4):213-228
  37. 37. Zaborowski MP, Balaj L, Breakefield XO, Lai CP. Extracellular vesicles: Composition, biological relevance, and methods of study. Bioscience. 2015;65(8):783-797
  38. 38. Chaudhary PK, Kim S, Kim S. Shedding light on the cell biology of platelet-derived extracellular vesicles and their biomedical applications. Life (Basel). 2023;13(6):1403
  39. 39. Wollert T, Hurley JH. Molecular mechanism of multivesicular body biogenesis by ESCRT complexes. Nature. 2010;464(7290):864-869
  40. 40. Trajkovic K, Hsu C, Chiantia S, Rajendran L, Wenzel D, Wieland F, et al. Ceramide triggers budding of exosome vesicles into multivesicular endosomes. Science. 2008;319(5867):1244-1247
  41. 41. Sinha A, Ignatchenko V, Ignatchenko A, Mejia-Guerrero S, Kislinger T. In-depth proteomic analyses of ovarian cancer cell line exosomes reveals differential enrichment of functional categories compared to the NCI 60 proteome. Biochemical and Biophysical Research Communications. 2014;445(4):694-701
  42. 42. Mobius W, Ohno-Iwashita Y, van Donselaar EG, Oorschot VM, Shimada Y, Fujimoto T, et al. Immunoelectron microscopic localization of cholesterol using biotinylated and non-cytolytic perfringolysin O. The Journal of Histochemistry and Cytochemistry. 2002;50(1):43-55
  43. 43. Simons M, Raposo G. Exosomes--vesicular carriers for intercellular communication. Current Opinion in Cell Biology. 2009;21(4):575-581
  44. 44. Chen H, Wang L, Zeng X, Schwarz H, Nanda HS, Peng X, et al. Exosomes, a new star for targeted delivery. Frontiers in Cell and Development Biology. 2021;9:751079
  45. 45. Cai H, Reinisch K, Ferro-Novick S. Coats, tethers, rabs, and SNARES work together to mediate the intracellular destination of a transport vesicle. Developmental Cell. 2007;12(5):671-682
  46. 46. Zhang W, Chen S, Liu ML. Pathogenic roles of microvesicles in diabetic retinopathy. Acta Pharmacologica Sinica. 2018;39(1):1-11
  47. 47. Hristov M, Erl W, Linder S, Weber PC. Apoptotic bodies from endothelial cells enhance the number and initiate the differentiation of human endothelial progenitor cells in vitro. Blood. 2004;104(9):2761-2766
  48. 48. Thery C, Boussac M, Veron P, Ricciardi-Castagnoli P, Raposo G, Garin J, et al. Proteomic analysis of dendritic cell-derived exosomes: A secreted subcellular compartment distinct from apoptotic vesicles. Journal of Immunology. 2001;166(12):7309-7318
  49. 49. Doyle LM, Wang MZ. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019;8(7):727
  50. 50. Hu W, Song X, Yu H, Sun J, Zhao Y. Therapeutic potentials of extracellular vesicles for the treatment of diabetes and diabetic complications. International Journal of Molecular Sciences. 2020;21(14):5163
  51. 51. van der Pol E, Boing AN, Harrison P, Sturk A, Nieuwland R. Classification, functions, and clinical relevance of extracellular vesicles. Pharmacological Reviews. 2012;64(3):676-705
  52. 52. Lv Y, Tan J, Miao Y, Zhang Q. The role of microvesicles and its active molecules in regulating cellular biology. Journal of Cellular and Molecular Medicine. 2019;23(12):7894-7904
  53. 53. Sun F, Xu W, Qian H. The emerging role of extracellular vesicles in retinal diseases. American Journal of Translational Research. 2021;13(12):13227-13245
  54. 54. Helwa I, Cai J, Drewry MD, Zimmerman A, Dinkins MB, Khaled ML, et al. A comparative study of serum exosome isolation using differential ultracentrifugation and three commercial reagents. PLoS One. 2017;12(1):e0170628
  55. 55. Massoumi H, Amin S, Soleimani M, Momenaei B, Ashraf MJ, Guaiquil VH, et al. Extracellular-vesicle-based therapeutics in neuro-ophthalmic disorders. International Journal of Molecular Sciences. 2023;24(10):9006
  56. 56. Zoller M. Tetraspanins: Push and pull in suppressing and promoting metastasis. Nature Reviews: Cancer. 2009;9(1):40-55
  57. 57. Zhu L, Zang J, Liu B, Yu G, Hao L, Liu L, et al. Oxidative stress-induced RAC autophagy can improve the HUVEC functions by releasing exosomes. Journal of Cellular Physiology. 2020;235(10):7392-7409
  58. 58. Jung MK, Mun JY. Sample preparation and imaging of exosomes by transmission electron microscopy. Journal of Visualized Experiments. 2018;131:56482
  59. 59. Ibrahim SA, Khan YS. Histology, extracellular vesicles. In: StatPearls. Treasure Island (Florida) ineligible companies. Disclosure: Yusuf Khan declares no relevant financial relationships with ineligible companies; 2024
  60. 60. Tokarz A, Szuscik I, Kusnierz-Cabala B, Kapusta M, Konkolewska M, Zurakowski A, et al. Extracellular vesicles participate in the transport of cytokines and angiogenic factors in diabetic patients with ocular complications. Folia Medica Cracoviensia. 2015;55(4):35-48
  61. 61. Elmasry K, Mohamed R, Sharma I, Elsherbiny NM, Liu Y, Al-Shabrawey M, et al. Epigenetic modifications in hyperhomocysteinemia: Potential role in diabetic retinopathy and age-related macular degeneration. Oncotarget. 2018;9(16):12562-12590
  62. 62. Bastiaans J, van Meurs JC, Mulder VC, Nagtzaam NM, Smits-te Nijenhuis M, Dufour-van den Goorbergh DC, et al. The role of thrombin in proliferative vitreoretinopathy. Investigative Ophthalmology & Visual Science. 2014;55(7):4659-4666
  63. 63. Vitkova V, Zivny J, Janota J. Endothelial cell-derived microvesicles: Potential mediators and biomarkers of pathologic processes. Biomarkers in Medicine. 2018;12(2):161-175
  64. 64. Huang H, Gandhi JK, Zhong X, Wei Y, Gong J, Duh EJ, et al. TNFalpha is required for late BRB breakdown in diabetic retinopathy, and its inhibition prevents leukostasis and protects vessels and neurons from apoptosis. Investigative Ophthalmology & Visual Science. 2011;52(3):1336-1344
  65. 65. Jansen F, Yang X, Franklin BS, Hoelscher M, Schmitz T, Bedorf J, et al. High glucose condition increases NADPH oxidase activity in endothelial microparticles that promote vascular inflammation. Cardiovascular Research. 2013;98(1):94-106
  66. 66. Ke Y, Fan X, Rui H, Xinjun R, Dejia W, Chuanzhen Z, et al. Exosomes derived from RPE cells under oxidative stress mediate inflammation and apoptosis of normal RPE cells through Apaf1/caspase-9 axis. Journal of Cellular Biochemistry. 2020;121(12):4849-4861
  67. 67. Tokarz A, Konkolewska M, Kusnierz-Cabala B, Maziarz B, Hanarz P, Zurakowski A, et al. Retinopathy severity correlates with RANTES concentrations and CCR 5-positive microvesicles in diabetes. Folia Medica Cracoviensia. 2019;59(3):95-112
  68. 68. Jansen F, Wang H, Przybilla D, Franklin BS, Dolf A, Pfeifer P, et al. Vascular endothelial microparticles-incorporated microRNAs are altered in patients with diabetes mellitus. Cardiovascular Diabetology. 2016;15:49
  69. 69. Zhang W, Dong X, Wang T, Kong Y. Exosomes derived from platelet-rich plasma mediate hyperglycemia-induced retinal endothelial injury via targeting the TLR4 signaling pathway. Experimental Eye Research. 2019;189:107813
  70. 70. Zhang W, Jiang H, Kong Y. Exosomes derived from platelet-rich plasma activate YAP and promote the fibrogenic activity of muller cells via the PI3K/Akt pathway. Experimental Eye Research. 2020;193:107973
  71. 71. Arroba AI, Valverde AM. Modulation of microglia in the retina: New insights into diabetic retinopathy. Acta Diabetologica. 2017;54(6):527-533
  72. 72. Madeira MH, Boia R, Santos PF, Ambrosio AF, Santiago AR. Contribution of microglia-mediated neuroinflammation to retinal degenerative diseases. Mediators of Inflammation. 2015;2015:673090
  73. 73. Aires ID, Ribeiro-Rodrigues T, Boia R, Catarino S, Girao H, Ambrosio AF, et al. Exosomes derived from microglia exposed to elevated pressure amplify the neuroinflammatory response in retinal cells. Glia. 2020;68(12):2705-2724
  74. 74. Beltramo E, Mazzeo A, Porta M. Release of pro-inflammatory/angiogenic factors by retinal microvascular cells is mediated by extracellular vesicles derived from M1-activated microglia. International Journal of Molecular Sciences. 2023;25(1):15
  75. 75. Liu Y, Yang Q , Fu H, Wang J, Yuan S, Li X, et al. Muller glia-derived exosomal miR-9-3p promotes angiogenesis by restricting sphingosine-1-phosphate receptor S1P(1) in diabetic retinopathy. Molecular Therapy - Nucleic Acids. 2022;27:491-504
  76. 76. Merle NS, Church SE, Fremeaux-Bacchi V, Roumenina LT. Complement system part I—Molecular mechanisms of activation and regulation. Frontiers in Immunology. 2015;6:262
  77. 77. Merle NS, Noe R, Halbwachs- Mecarelli L, Fremeaux-Bacchi V, Roumenina LT. Complement system part II: Role in immunity. Frontiers in Immunology. 2015;6:257
  78. 78. Gerl VB, Bohl J, Pitz S, Stoffelns B, Pfeiffer N, Bhakdi S. Extensive deposits of complement C3d and C5b-9 in the choriocapillaris of eyes of patients with diabetic retinopathy. Investigative Ophthalmology & Visual Science. 2002;43(4):1104-1108
  79. 79. Zhang J, Gerhardinger C, Lorenzi M. Early complement activation and decreased levels of glycosylphosphatidylinositol-anchored complement inhibitors in human and experimental diabetic retinopathy. Diabetes. 2002;51(12):3499-3504
  80. 80. Huang C, Fisher KP, Hammer SS, Busik JV. Extracellular vesicle-induced classical complement activation leads to retinal endothelial cell damage via MAC deposition. International Journal of Molecular Sciences. 2020;21(5):1693
  81. 81. Huang C, Fisher KP, Hammer SS, Navitskaya S, Blanchard GJ, Busik JV. Plasma exosomes contribute to microvascular damage in diabetic retinopathy by activating the classical complement pathway. Diabetes. 2018;67(8):1639-1649
  82. 82. Atienzar-Aroca S, Flores-Bellver M, Serrano-Heras G, Martinez-Gil N, Barcia JM, Aparicio S, et al. Oxidative stress in retinal pigment epithelium cells increases exosome secretion and promotes angiogenesis in endothelial cells. Journal of Cellular and Molecular Medicine. 2016;20(8):1457-1466
  83. 83. Wu SF, Noren Hooten N, Freeman DW, Mode NA, Zonderman AB, Evans MK. Extracellular vesicles in diabetes mellitus induce alterations in endothelial cell morphology and migration. Journal of Translational Medicine. 2020;18(1):230
  84. 84. Mazzeo A, Beltramo E, Lopatina T, Gai C, Trento M, Porta M. Molecular and functional characterization of circulating extracellular vesicles from diabetic patients with and without retinopathy and healthy subjects. Experimental Eye Research. 2018;176:69-77
  85. 85. Xiao J, Zhang H, Yang F, Xiao M, Zhou L, Yu R, et al. Proteomic analysis of plasma sEVs reveals that TNFAIP8 is a new biomarker of cell proliferation in diabetic retinopathy. Journal of Proteome Research. 2021;20(3):1770-1782
  86. 86. Martins B, Amorim M, Reis F, Ambrosio AF, Fernandes R. Extracellular vesicles and MicroRNA: Putative role in diagnosis and treatment of diabetic retinopathy. Antioxidants (Basel). 2020;9(8):705
  87. 87. Wang P, Li C, Deng Y, Yu Q , Meng X, Jiang T, et al. Effect of plasma-derived extracellular vesicles on angiogenesis and the ensuing proliferative diabetic retinopathy through a miR-30b-dependent mechanism. Diabetology and Metabolic Syndrome. 2022;14(1):188
  88. 88. Mishra M, Duraisamy AJ, Kowluru RA. Sirt1: A guardian of the development of diabetic retinopathy. Diabetes. 2018;67(4):745-754
  89. 89. Murinello S, Usui Y, Sakimoto S, Kitano M, Aguilar E, Friedlander HM, et al. miR-30a-5p inhibition promotes interaction of Fas(+) endothelial cells and FasL(+) microglia to decrease pathological neovascularization and promote physiological angiogenesis. Glia. 2019;67(2):332-344
  90. 90. Gong M, Yu B, Wang J, Wang Y, Liu M, Paul C, et al. Mesenchymal stem cells release exosomes that transfer miRNAs to endothelial cells and promote angiogenesis. Oncotarget. 2017;8(28):45200-45212
  91. 91. Chen Q , Qiu F, Zhou K, Matlock HG, Takahashi Y, Rajala RVS, et al. Pathogenic role of microRNA-21 in diabetic retinopathy through downregulation of PPARalpha. Diabetes. 2017;66(6):1671-1682
  92. 92. Mazzeo A, Lopatina T, Gai C, Trento M, Porta M, Beltramo E. Functional analysis of miR-21-3p, miR-30b-5p and miR-150-5p shuttled by extracellular vesicles from diabetic subjects reveals their association with diabetic retinopathy. Experimental Eye Research. 2019;184:56-63
  93. 93. Liu CH, Sun Y, Li J, Gong Y, Tian KT, Evans LP, et al. Endothelial microRNA-150 is an intrinsic suppressor of pathologic ocular neovascularization. Proceedings of the National Academy of Sciences of the United States of America. 2015;112(39):12163-12168
  94. 94. Beltramo E, Porta M. Pericyte loss in diabetic retinopathy: Mechanisms and consequences. Current Medicinal Chemistry. 2013;20(26):3218-3225
  95. 95. Liu C, Ge HM, Liu BH, Dong R, Shan K, Chen X, et al. Targeting pericyte-endothelial cell crosstalk by circular RNA-cPWWP2A inhibition aggravates diabetes-induced microvascular dysfunction. Proceedings of the National Academy of Sciences of the United States of America. 2019;116(15):7455-7464
  96. 96. Lee J, Kim KE, Choi DK, Jang JY, Jung JJ, Kiyonari H, et al. Angiopoietin-1 guides directional angiogenesis through integrin alphavbeta5 signaling for recovery of ischemic retinopathy. Science Translational Medicine. 2013;5(203):203ra127
  97. 97. Chahed S, Leroyer AS, Benzerroug M, Gaucher D, Georgescu A, Picaud S, et al. Increased vitreous shedding of microparticles in proliferative diabetic retinopathy stimulates endothelial proliferation. Diabetes. 2010;59(3):694-701
  98. 98. Maisto R, Trotta MC, Petrillo F, Izzo S, Cuomo G, Alfano R, et al. Resolvin D1 modulates the intracellular VEGF-related miRNAs of retinal photoreceptors challenged with high glucose. Frontiers in Pharmacology. 2020;11:235
  99. 99. Maisto R, Oltra M, Vidal-Gil L, Martinez-Gil N, Sancho-Pelluz J, Filippo CD, et al. ARPE-19-derived VEGF-containing exosomes promote neovascularization in HUVEC: The role of the melanocortin receptor 5. Cell Cycle. 2019;18(4):413-424
  100. 100. Stepien E, Kablak-Ziembicka A, Czyz J, Przewlocki T, Malecki M. Microparticles, not only markers but also a therapeutic target in the early stage of diabetic retinopathy and vascular aging. Expert Opinion on Therapeutic Targets. 2012;16(7):677-688
  101. 101. Ratajczak MZ. The emerging role of microvesicles in cellular therapies for organ/tissue regeneration. Nephrology, Dialysis, Transplantation. 2011;26(5):1453-1456
  102. 102. Beltramo E, Lopatina T, Berrone E, Mazzeo A, Iavello A, Camussi G, et al. Extracellular vesicles derived from mesenchymal stem cells induce features of diabetic retinopathy in vitro. Acta Diabetologica. 2014;51(6):1055-1064
  103. 103. Kamalden TA, Macgregor-Das AM, Kannan SM, Dunkerly-Eyring B, Khaliddin N, Xu Z, et al. Exosomal microRNA-15a transfer from the pancreas augments diabetic complications by inducing oxidative stress. Antioxidants & Redox Signaling. 2017;27(13):913-930
  104. 104. Razmara M, Hjemdahl P, Ostenson CG, Li N. Platelet hyperprocoagulant activity in type 2 diabetes mellitus: Attenuation by glycoprotein IIb/IIIa inhibition. Journal of Thrombosis and Haemostasis. 2008;6(12):2186-2192
  105. 105. Su Y, Chen J, Dong Z, Zhang Y, Ma R, Kou J, et al. Procoagulant activity of blood and endothelial cells via phosphatidylserine exposure and microparticle delivery in patients with diabetic retinopathy. Cellular Physiology and Biochemistry. 2018;45(6):2411-2420
  106. 106. Zhang X, McGeoch SC, Johnstone AM, Holtrop G, Sneddon AA, MacRury SM, et al. Platelet-derived microparticle count and surface molecule expression differ between subjects with and without type 2 diabetes, independently of obesity status. Journal of Thrombosis and Thrombolysis. 2014;37(4):455-463
  107. 107. Li X, Yu ZW, Wang Y, Fu YH, Gao XY. MicroRNAs: Potential targets in diabetic retinopathy. Hormone and Metabolic Research. 2020;52(3):142-148
  108. 108. Zhuang X, Ji L, Jiang H, Liu Y, Liu X, Bi J, et al. Exosomes derived from stem cells from the apical papilla promote dentine-pulp complex regeneration by inducing specific dentinogenesis. Stem Cells International. 2020;2020:5816723
  109. 109. Zhang Y, Wei J, Zhang L, Jiang G, Wang B, Jiang L. Extracellular vesicle-derived miR-26b-5p is up-regulated in the serum of patients with diabetic retinopathy. Combinatorial Chemistry & High Throughput Screening. 2022;25(5):877-882
  110. 110. Mighty J, Rubio-Navarro A, Shi C, Zhou J, Flores-Bellver M, Heissel S, et al. Extracellular vesicles of human diabetic retinopathy retinal tissue and urine of diabetic retinopathy patients are enriched for the junction plakoglo bin protein. Frontiers in Endocrinology (Lausanne). 2022;13:1077644
  111. 111. Qing S, Yuan S, Yun C, Hui H, Mao P, Wen F, et al. Serum miRNA biomarkers serve as a fingerprint for proliferative diabetic retinopathy. Cellular Physiology and Biochemistry. 2014;34(5):1733-1740
  112. 112. Friedrich J, Steel DHW, Schlingemann RO, Koss MJ, Hammes HP, Krenning G, et al. microRNA expression profile in the vitreous of proliferative diabetic retinopathy patients and differences from patients treated with anti-VEGF therapy. Translational Vision Science & Technology. 2020;9(6):16
  113. 113. Qin LL, An MX, Liu YL, Xu HC, Lu ZQ. MicroRNA-126: A promising novel biomarker in peripheral blood for diabetic retinopathy. International Journal of Ophthalmology. 2017;10(4):530-534
  114. 114. Markomichelakis NN, Theodossiadis PG, Sfikakis PP. Regression of neovascular age-related macular degeneration following infliximab therapy. American Journal of Ophthalmology 2005;139(3):537-540
  115. 115. Stahel M, Becker M, Graf N, Michels S. Systemic interleukin 1beta inhibition in proliferative diabetic retinopathy: A prospective open-label study using canakinumab. Retina. 2016;36(2):385-391
  116. 116. Rodriguez ML, Perez S, Mena-Molla S, Desco MC, Ortega AL. Oxidative stress and microvascular alterations in diabetic retinopathy: Future therapies. Oxidative Medicine and Cellular Longevity. 2019;2019:4940825
  117. 117. Habibi A, Zarei-Behjani Z, Falamarzi K, Malekpour M, Ebrahimi F, Soleimani M, et al. Extracellular vesicles as a new horizon in the diagnosis and treatment of inflammatory eye diseases: A narrative review of the literature. Frontiers in Immunology. 2023;14:1097456
  118. 118. Safwat A, Sabry D, Ragiae A, Amer E, Mahmoud RH, Shamardan RM. Adipose mesenchymal stem cells-derived exosomes attenuate retina degeneration of streptozotocin-induced diabetes in rabbits. Journal of Circulating Biomarkers. 2018;7:1849454418807827
  119. 119. Zhang W, Wang Y, Kong Y. Exosomes derived from mesenchymal stem cells modulate miR-126 to ameliorate hyperglycemia-induced retinal inflammation via targeting HMGB1. Investigative Ophthalmology & Visual Science. 2019;60(1):294-303
  120. 120. Gu C, Zhang H, Gao Y. Adipose mesenchymal stem cells-secreted extracellular vesicles containing microRNA-192 delays diabetic retinopathy by targeting ITGA1. Journal of Cellular Physiology. 2021;236(7):5036-5051
  121. 121. Reddy SK, Ballal AR, Shailaja S, Seetharam RN, Raghu CH, Sankhe R, et al. Small extracellular vesicle-loaded bevacizumab reduces the frequency of intravitreal injection required for diabetic retinopathy. Theranostics. 2023;13(7):2241-2255
  122. 122. Sun F, Sun Y, Zhu J, Wang X, Ji C, Zhang J, et al. Mesenchymal stem cells-derived small extracellular vesicles alleviate diabetic retinopathy by delivering NEDD4. Stem Cell Research & Therapy. 2022;13(1):293
  123. 123. Hajrasouliha AR, Jiang G, Lu Q , Lu H, Kaplan HJ, Zhang HG, et al. Exosomes from retinal astrocytes contain antiangiogenic components that inhibit laser-induced choroidal neovascularization. The Journal of Biological Chemistry. 2013;288(39):28058-28067
  124. 124. Gu S, Liu Y, Zou J, Wang W, Wei T, Wang X, et al. Retinal pigment epithelial cells secrete miR-202-5p-containing exosomes to protect against proliferative diabetic retinopathy. Experimental Eye Research. 2020;201:108271
  125. 125. Huang S, Zeng Y, Guo Q , Zou T, Yin ZQ. Small extracellular vesicles of organoid-derived human retinal stem cells remodel Muller cell fate via miRNA: A novel remedy for retinal degeneration. Journal of Controlled Release. 2024;370:405-420
  126. 126. Yang C, Xiong W, Qiu Q , Shao Z, Hamel D, Tahiri H, et al. Role of receptor-mediated endocytosis in the antiangiogenic effects of human T lymphoblastic cell-derived microparticles. American Journal of Physiology. Regulatory, Integrative and Comparative Physiology. 2012;302(8):R941-R949
  127. 127. Lamb WDB, Eastlake K, Luis J, Sharif NA, Khaw PT, Limb GA. MicroRNA profile of extracellular vesicles released by Muller glial cells. Frontiers in Cellular Neuroscience. 2023;17:1325114
  128. 128. Mathew B, Acha LG, Torres LA, Huang CC, Liu A, Kalinin S, et al. MicroRNA-based engineering of mesenchymal stem cell extracellular vesicles for treatment of retinal ischemic disorders: Engineered extracellular vesiclesand retinal ischemia. Acta Biomaterialia. 2023;158:782-797
  129. 129. Shaker OG, Abdelaleem OO, Mahmoud RH, Abdelghaffar NK, Ahmed TI, Said OM, et al. Diagnostic and prognostic role of serum miR-20b, miR-17-3p, HOTAIR, and MALAT1 in diabetic retinopathy. IUBMB Life. 2019;71(3):310-320
  130. 130. Li Z, Dong Y, He C, Pan X, Liu D, Yang J, et al. RNA-Seq revealed novel non-proliferative retinopathy specific circulating MiRNAs in T2DM patients. Frontiers in Genetics. 2019;10:531
  131. 131. McArthur K, Feng B, Wu Y, Chen S, Chakrabarti S. MicroRNA-200b regulates vascular endothelial growth factor-mediated alterations in diabetic retinopathy. Diabetes. 2011;60(4):1314-1323
  132. 132. Blum A, Meerson A, Rohana H, Jabaly H, Nahul N, Celesh D, et al. MicroRNA-423 may regulate diabetic vasculopathy. Clinical and Experimental Medicine. 2019;19(4):469-477
  133. 133. Sun F, Sun Y, Wang X, Zhu J, Chen S, Yu Y, et al. Engineered mesenchymal stem cell-derived small extracellular vesicles for diabetic retinopathy therapy through HIF-1α/EZH2/PGC-1α pathway. Bioactive Materials. 2024;33:444-459
  134. 134. Haraszti RA, Miller R, Stoppato M, Sere YY, Coles A, Didiot MC, et al. Exosomes produced from 3D cultures of MSCs by tangential flow filtration show higher yield and improved activity. Molecular Therapy. 2018;26(12):2838-2847
  135. 135. Lener T, Gimona M, Aigner L, Borger V, Buzas E, Camussi G, et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. Journal of Extracellular Vesicles. 2015;4:30087
  136. 136. Zhao K, Lou R, Huang F, Peng Y, Jiang Z, Huang K, et al. Immunomodulation effects of mesenchymal stromal cells on acute graft-versus-host disease after hematopoietic stem cell transplantation. Biology of Blood and Marrow Transplantation. 2015;21(1):97-104
  137. 137. Shelke GV, Lasser C, Gho YS, Lotvall J. Importance of exosome depletion protocols to eliminate functional and RNA-containing extracellular vesicles from fetal bovine serum. Journal of Extracellular Vesicles. 2014:3(1):24783
  138. 138. Pachler K, Lener T, Streif D, Dunai ZA, Desgeorges A, Feichtner M, et al. A good manufacturing practice-grade standard protocol for exclusively human mesenchymal stromal cell-derived extracellular vesicles. Cytotherapy. 2017;19(4):458-472
  139. 139. Nordin JZ, Lee Y, Vader P, Mager I, Johansson HJ, Heusermann W, et al. Ultrafiltration with size-exclusion liquid chromatography for high yield isolation of extracellular vesicles preserving intact biophysical and functional properties. Nanomedicine. 2015;11(4):879-883
  140. 140. Benedikter BJ, Bouwman FG, Vajen T, Heinzmann ACA, Grauls G, Mariman EC, et al. Ultrafiltration combined with size exclusion chromatography efficiently isolates extracellular vesicles from cell culture media for compositional and functional studies. Scientific Reports. 2017;7(1):15297
  141. 141. Lorincz AM, Timar CI, Marosvari KA, Veres DS, Otrokocsi L, Kittel A, et al. Effect of storage on physical and functional properties of extracellular vesicles derived from neutrophilic granulocytes. Journal of Extracellular Vesicles. 2014;3:25465
  142. 142. Larson MC, Luthi MR, Hogg N, Hillery CA. Calcium-phosphate microprecipitates mimic microparticles when examined with flow cytometry. Cytometry. Part A. 2013;83(2):242-250
  143. 143. Kalra H, Adda CG, Liem M, Ang CS, Mechler A, Simpson RJ, et al. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics. 2013;13(22):3354-3364
  144. 144. Kusuma GD, Barabadi M, Tan JL, Morton DAV, Frith JE, Lim R. To protect and to preserve: Novel preservation strategies for extracellular vesicles. Frontiers in Pharmacology. 2018;9:1199
  145. 145. Yin H, Chen CY, Liu YW, Tan YJ, Deng ZL, Yang F, et al. Synechococcus elongatus PCC7942 secretes extracellular vesicles to accelerate cutaneous wound healing by promoting angiogenesis. Theranostics. 2019;9(9):2678-2693
  146. 146. Hu W, Song X, Yu H, Sun J, Zhao Y. Released exosomes contribute to the immune modulation of cord blood-derived stem cells. Frontiers in Immunology. 2020;11:165
  147. 147. Murphy DE, de Jong OG, Brouwer M, Wood MJ, Lavieu G, Schiffelers RM, et al. Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Experimental & Molecular Medicine. 2019;51(3):1-12

Written By

Samar Habib, Alaa M. Mansour, Babak Baban and Khaled Elmasry

Submitted: 17 February 2024 Reviewed: 23 April 2024 Published: 24 June 2024