Next Article in Journal
The Role of Alternative Mitophagy in Heart Disease
Next Article in Special Issue
Extracellular Vesicles Secreted by Pre-Hatching Bovine Embryos Produced In Vitro and In Vivo Alter the Expression of IFNtau-Stimulated Genes in Bovine Endometrial Cells
Previous Article in Journal
Towards Precision Prognostication and Personalized Therapeutics through Proteomics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Salivary Exosomes in Health and Disease: Future Prospects in the Eye

1
Texas College of Osteopathic Medicine, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
2
North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
3
Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
4
Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(7), 6363; https://doi.org/10.3390/ijms24076363
Submission received: 16 February 2023 / Revised: 21 March 2023 / Accepted: 24 March 2023 / Published: 28 March 2023
(This article belongs to the Special Issue Extracellular Vesicles: The Biology and Therapeutic Applications)

Abstract

:
Exosomes are a group of vesicles that package and transport DNA, RNA, proteins, and lipids to recipient cells. They can be derived from blood, saliva, urine, and/or other biological tissues. Their impact on several diseases, such as neurodegenerative, autoimmune, and ocular diseases, have been reported, but not fully unraveled. The exosomes that are derived from saliva are less studied, but offer significant advantages over exosomes from other sources, due to their accessibility and ease of collection. Thus, their role in the pathophysiology of diseases is largely unknown. In the context of ocular diseases, salivary exosomes have been under-utilized, thus creating an enormous gap in the literature. The current review discusses the state of exosomes research on systemic and ocular diseases and highlights the role and potential of salivary exosomes as future ocular therapeutic vehicles.

1. Introduction

Extracellular vesicles are membranous vesicles that are released from cells originating from the plasma membrane or endosomal system [1]. Currently, extracellular vesicles are broadly divided into three types: microvesicles, apoptotic vesicles, and exosomes [2,3,4]. Universal definitions for these three categories of extracellular vesicles do not currently exist, since the techniques to isolate and characterize extracellular vesicles properly are widely diverse and being optimized [5,6,7]. Additionally, there are size overlaps between the types of extracellular vesicles. Microvesicles, which are released from cells via the shedding of the plasma membrane, range from 100 to 1000 nm in diameter [8]. Exosomes range in size from 50 to 150 nm in diameter, making up the smallest type of extracellular vesicles [8], while apoptotic vesicles range in size from 1000 to 5000 nm in diameter [8,9]. Therefore, exosomes will henceforth be differentiated in this article by their relatively small size and further specified by their cell of origin. Exosomes are derived from endosomal membrane components and secreted after fusion with the plasma membrane [8,10], as shown in Figure 1A. After being released to the extracellular space and circulation, exosomes transfer their cargo into recipient target cells through endocytosis [11]. It is worth noting that other than the exosomes in blood circulation, most exosomes are not considered to be long-range effectors, as they are taken up by cells that are undirected in the immediate area of exosome release [11]. Through this interaction, exosomes can modify gene expression, cellular signaling, and the overall function of target cells [11]. Like their wide range of effects on recipient cells, the composition of exosomes is also diverse, containing markers including CD63, CD9, CD81, and TSG101, and essential biological molecules, such as DNA, RNA, proteins, and lipids. These components are integral to the roles of exosomes in intracellular communication and transport, as shown in Figure 1B [11,12,13,14]. Exosomes also carry IgA and plgR, which are anti-inflammatory biomarkers [15], highlighting their role in mediating the immune responses to inflammation, cell migration, and wound healing [16]. Exosomes are found in various body fluids, such as plasma [17,18,19], saliva [20,21,22], urine [23,24,25,26], and amniotic fluid [27,28,29]. Due to their versatility of function, they have gained traction over the last 20 years to serve as diagnostic biomarkers and potential treatments for numerous diseases.
Saliva offers several advantages over blood for clinical diagnosis [30,31,32]. Specifically, saliva collection is easy, non-invasive, cost-effective [31,33], does not coagulate, and is easier to handle [30,31,32]. Salivary exosomes (SEs) are being investigated as an alternative to whole saliva due to the contaminating elements and higher amylase enzyme levels that are found in whole saliva [15,34]. Additionally, the proteins and RNAs from whole saliva are susceptible to degradation when removed from their natural environment, so the protease and RNase enzymes in saliva must be stabilized with protease inhibitors and RNase inhibitors, in order to protect against the deterioration of salivary proteins and RNA [35,36,37]. Conversely, SEs, along with exosomes from other bodily fluids, have a lipid bilayer that protects their cargo, providing more accurate information for clinical diagnosis and treatment [30].
Compared to the exosomes from other bodily fluids, SEs are thought to offer a greater versatility in diagnosing and treating diseases [34,38,39]. For instance, the applications of urinary exosomes are limited to kidney and prostate pathologies, such as hyperaldosteronism [40] and prostate cancer [38,41]. On the other hand, SEs have applications in systemic diseases, in addition to organ-specific pathologies, since saliva has been reported to carry the DNA, RNA, and metabolites that are found in both the blood and saliva [30,34,39]. The advantages/disadvantages of SEs over other exosomes are summarized in Table 1.
A few drawbacks also exist with SEs [42,43]. One important factor is the inter- and intra-subject variability of the salivary flow rate and composition, depending on the method of sampling used and the subjects’ food intake, aging, and preexisting conditions [33]. Thus, the best protocol for isolating these SEs is still debatable [42,44]. A common limitation of both SEs and non-SEs is the lack of techniques to precisely quantify their molecular content [42,43,44]. Thus, understanding the components and function of exosomes in relation to pathologies is crucial to developing more disease-targeted therapies.
Due to their versatility, SEs have been studied as biomarkers for diagnosing systemic autoimmune diseases, such as oral lichen planus and inflammatory bowel disease [45,46,47]. Recently, SEs were found to carry proteins that may propagate neoplastic and neurodegenerative diseases [22,48,49,50]. However, little is known about the therapeutic effects of the SEs in ocular tissue. Here, we discuss and provide an overview of the involvement of non-SEs and SEs in systemic and ocular diseases. Each subsequent section will be organized as follows: starting with describing the pathophysiology of the disease, followed by a discussion of the non-SEs in the disease, and finally ending with a review of the SEs in the disease.

2. Exosomes in Non-Ocular Diseases

2.1. Systemic Autoimmune Diseases

The roles of exosomes in autoimmune diseases have been extensively studied. In this section, we will discuss the pathophysiology, ocular manifestations, and effects of exosomes in oral lichen planus, periodontitis, Sjogren’s syndrome, and inflammatory bowel disease [45,51,52,53], which are summarized in Table 2.

2.1.1. Oral Lichen Planus

Lichen planus is a chronic inflammatory disease that is caused by T-cell proliferation and the apoptosis of the keratinocytes within the skin and mucosal membranes [54]. Oral lichen planus (OLP) occurs when inflammatory cells attack the keratinocytes within the oral mucosal lining, resulting in erythematous ulcers [55,56]. Although more rare than OLP, lichen planus may also involve ocular structures such as the eyelids, cornea, and conjunctiva, manifesting as blepharitis [57], keratitis [58], corneal perforation [59], and cicatrizing conjunctivitis [54,60,61]. To date, no exosomal studies on conjunctival lichen planus have been reported, but there are numerous reports on exosomes and oral lichen planus [62,63,64]. Plasma-derived exosomes from OLP patients were found to enhance T-cell proliferation and migration, and to upregulate pro-inflammatory cytokines such as interferon-gamma (IFN-γ), accelerating OLP progression [65]. In these studies, the plasma exosome microRNA, (miR)-301b-3p, was downregulated, while miR-130b-3p and miR-34a-5p were upregulated [62]. Interestingly, miR-34a-5p was found to be positively correlated with the severity of the clinical symptoms of OLP [62]. Yang and co-authors observed that T-cell-derived exosomes from OLP patients stimulated the production of macrophage inflammatory protein-1 alpha/beta and promoted the migration of CD8+ T cells to the OLP lesions [64]. These exosomes were later found to trigger the apoptosis of keratinocytes in vitro, thus contributing to the exacerbation of OLP [63].
Several studies have suggested the roles of SEs in modulating the gene expression in OLP [15,45,46]. In vitro, fluorescently labeled SEs are taken up by oral keratinocytes and subsequently modulate the oral keratinocytes’ gene expression, by transferring mRNA to the keratinocytes [15]. Besides in vitro studies, the utility of SEs in transporting the miRNAs within humans has also been demonstrated [45]. In 16 OLP patients and 8 healthy controls, Byun et al. revealed that miR-4484 in the SEs derived from OLP patients was significantly upregulated, relative to the healthy controls, indicating that miR-4484 may provide insight into the pathogenic mechanisms of OLP [45].

2.1.2. Periodontitis

Periodontitis is an inflammation of gingival tissue that is characterized by microbial dysbiosis in the oral cavity and a subsequent loss of periodontal ligament attachment and alveolar bone [51,66]. Due to the shared risk factors between periodontitis and various ocular diseases, periodontitis has been shown to be correlated with an increased risk of developing primary open-angle glaucoma [67], cataracts [68,69], scleritis [70], and age-related macular degeneration [71,72]. Non-SEs have exhibited both pro-inflammatory and immunoprotective effects in periodontitis [73,74]. Zhang et al. showed that isolated periodontal ligament stem cell (PDLSCs) exosomes upregulated the inflammation and vascularization of the periodontal ligaments via the transfer of vascular endothelial growth factor (VEGF) [73]. However, the overexpression of exosomal miR-17-5p, an inhibitor of VEGF, slowed the vascularization of the inflamed PDLSCs [73]. Conversely, Shen et al. demonstrated the therapeutic effect of exosomes that were derived from dental pulp stem cells, showing the acceleration of alveolar bone and periodontal epithelium healing in mice with periodontitis [74]. The suppression of periodontal inflammation, by converting the macrophages in the periodontium from pro-inflammatory to anti-inflammatory phenotypes, was also shown [74].
SEs in periodontitis have garnered recent interest [51,75]. Notably, the SEs from periodontitis patients contained higher programmed death-ligand 1 (PD-L1) levels compared to healthy controls, which is positively correlated to the disease severity [75]. Han et al. studied the diagnostic potential of ten miRNAs that were expressed in the SEs, compared to whole saliva that was derived from patients with periodontitis or gingivitis, as well as from healthy individuals [51]. A total of three of those miRNAs (miR-140-5p, miR-146a-5p, and miR-628-5p) were significantly increased in the periodontitis patients, relative to the healthy controls, and were only detected in the SEs from the periodontitis patients, compared to whole saliva, as shown in Figure 2 [51]. When isolated from the whole saliva, miR-140-5p, miR-146a-5p, and miR-628-5p exhibited a low discriminatory power (AUC of 0.57, 0.56, and 0.73, in Figure 2a,d,g) [51]. In contrast, when isolated from the SEs, miR-140-5p, miR-146a-5p, and miR-628-5p demonstrated a high discriminatory power, as indicated by the area under the curve (AUC) of 1, 0.97, and 0.93 (Figure 2b,e,h), highlighting the potential utility of miRNAs in SEs [51].
A recent study compared periodontitis patients, gingivitis patients, and healthy individuals by looking at lipopolysaccharide, a virulence factor that is expressed on the outer membrane of Gram-negative bacteria, and the global DNA methylation patterns of the 5-methylcytosine (5mC), 5-hydroxymethylcytosine (5hmC), and N6-Methyladenosine (m6dA) in SEs and whole saliva [76]. The LPS and global 5mC methylation were significantly upregulated in the periodontitis patients compared to the healthy controls, and these differences were more striking in the SEs than in the whole saliva [76]. Additionally, global 5mC hypermethylation could discriminate between periodontitis and gingivitis with a high sensitivity and specificity, as indicated by an AUC of 1 (Figure 3b), suggesting that the global methylation in SEs may be a biomarker for periodontal disease [76]. Together, these data suggest that, compared to whole saliva, SEs represent a source of biomarkers with an improved diagnostic potential for periodontal disease.

2.1.3. Sjogren’s Syndrome

Sjogren’s syndrome (SS) is a chronic autoimmune disorder in which lymphocytic infiltrates trigger the inflammation of exocrine glands [77,78]. The common clinical presentations of SS include keratoconjunctivitis sicca (dry eye) and xerostomia (dry mouth) [77,78]. SS has been associated with an increased risk of vision-threatening ocular morbidities, including optic neuropathy, uveitis, episcleritis, retinal vasculitis, corneal perforation, and cicatrizing conjunctivitis [79]. Since it has been shown that exosomes transport the miRNAs that are involved in autoimmunity and the immune response, the role of exosomes in SS has been explored extensively [52,80,81]. Recent studies have revealed that exosomes that were secreted by T cells derived from SS patients contained higher concentrations of miR-142-3p [81], which has been shown to contribute to the pathogenesis of SS through a decreased cAMP production, calcium signaling, and the subsequent reduced protein production of the salivary glands [81].
Several recent studies have suggested immunoprotective roles for exosomes in SS [52,80]. Using a rabbit model of autoimmune dry eye, Li et al. reported that the exosomes from human umbilical-cord-derived mesenchymal stem cells (MSCs) alleviated ophthalmitis by polarizing peripheral blood macrophages toward the anti-inflammatory M2 phenotype [82]. More recently, the exosomes from labial-gland-derived MSCs were found to enhance the salivary gland function in non-obese mice by decreasing the inflammatory infiltrates in the salivary glands [80]. An intravenous injection of these MSC-derived exosomes stimulated the proliferation of regulatory T cells in SS patients while blocking the production of T-helper 17 (Th17) cells [80]. Similarly, in an in vitro co-culture system of human umbilical cord MSCs and spleen mononuclear cells that were isolated from non-obese diabetic mice, the MSCs downregulated the expression of the pro-inflammatory cytokines (IFN-γ, IL-6, and tumor necrosis factor [TNF]-α) in mononuclear cells [83]. Tested in SS patients, the MSCs induced similar suppressive effects on the T cells, upregulating anti-inflammatory factor IL-10 by nearly three-fold (70 ± 5 pg/mL vs. 18 ± 3 pg/mL) and the regulatory T cells by more than two-fold (6.8 ± 1.6% vs. 3.3 ± 0.6% of total T cells), relative to the healthy controls [83]. Rui et al. demonstrated the therapeutic potential of exosomes that were derived from murine olfactory ecto-b stem cells, which upregulated the immunosuppressive effects of myeloid-derived suppressor cells (MDSCs) and slowed the SS progression, as measured by the increased saliva flow rates and reduced lymphocyte infiltration in the submandibular glands [52]. Specifically, S100A4, a ligand of toll-like receptor (TLR) 4 that is secreted by exosomes, modulated the secretion of IL-6 by MDSCs via TLR4 signaling [52].
Like non-SEs, SEs have been investigated in SS as potential biomarkers and therapeutic targets that can alleviate xerostomia in these patients [20,84,85]. Michael et al. were the first to isolate exosomal miRNAs from the saliva of SS patients [20]. They found that hsa-miR-23a was highly expressed in the SS patients but not in their healthy counterparts, suggesting that the miRNA content of SEs could provide novel markers for the diagnosis of various salivary gland diseases, including SS [20]. Hu et al. found that the expression of 16 peptides and 27 mRNAs was significantly modified in the SS patients compared to the healthy controls [86]. Alevizos et al. also examined the miRNAs that were carried by the SEs and discovered that miR-768-3p and miR-574-3p could identify the SS patients based on their salivary gland focus scores, which rate the intensity of the inflammatory lymphocytic infiltrates [85]. Gallo et al. found that the SEs in SS patients could serve as vehicles for transporting the miRNA that is expressed by the Epstein–Barr virus (ebv-miR-BART13-3p), from B cells to salivary epithelial cells [84].
In addition to miRNA transport, the exosomes from salivary gland epithelial cells carry the autoantigens anti-Ro/SSA, anti-La/SSB, and Sm ribonucleoproteins, suggesting that SEs may mediate the autoantigen presentation of lymphocytes in the progression of SS [87]. Aqrawi et al. used a liquid chromatograph-mass spectrometer (LC-MS) to analyze the proteomic biomarkers in the whole saliva and SEs from SS patients and healthy controls. The study found that the proteins that are involved in innate immunity and wound repair were upregulated in the saliva of the SS patients, when compared to the healthy cohort [88]. Interestingly, neutrophil gelatinase-associated lipocalin, an iron-binding protein that activates neutrophils in the innate immune system, was the most upregulated protein in the SEs of SS patients [88].

2.1.4. Inflammatory Bowel Disease

Inflammatory bowel disease (IBD) is an autoimmune disorder that is characterized by chronic inflammation and ulcers along the colon and rectum [89,90,91]. The common ocular manifestations of IBD include episcleritis [92,93,94,95] and uveitis [94,95,96,97]. Females have been reported to be at a significant risk for developing such ocular manifestations [96,98,99].
The exosomes secreted by MSCs have demonstrated immunoprotective effects in IBD [100,101,102]. For instance, the exosomes from human umbilical cord MSCs have been shown to alleviate the colon tissue inflammation in IBD mice via the upregulation of IL-10 and downregulation of TNF-α, IL-6, and IL-1β [100,103]. The suggested mechanisms for the exosomal protection of colon tissue from damage include suppressing macrophage infiltration into colon tissues [103] and blocking ubiquitination [100], which has been known to contribute to the development of IBD [104,105]. A recent study demonstrated the role of MSC-derived exosomes in inhibiting ubiquitination by transferring miR-326, leading to the downregulation of the pro-inflammatory nuclear factor-kappaB (NF-κB) signaling pathway [106]. More recently, it has been reported that MSC-derived exosomes provided protection against IBD damage by upregulating the TNF-α stimulated gene 6 in mice [101]. In addition, exosomes from human adipose MSCs promoted the regeneration of intestinal stem cells and epithelial cells [102].
Exosomes from sources other than MSCs have also been shown to be able to accelerate and attenuate IBD progression [53,107,108]. Wong et al. used dextran sulfate to simulate acute colitis in a mouse model and examine the role of serum exosomes in the macrophage activation of IBD [107]. The 56 differentially expressed proteins in the exosomes from the acute colitis mice were mainly involved in acute-phase proteins and the immunoglobulins that are involved in the coagulation cascade, which has been associated with macrophage activation [107]. Liao et al. showed that exosomes that were secreted from T regulatory cells relieved IBD by transporting miR-195a-3p to colonic epithelial cells [108]. In contrast, macrophage-derived exosomes that carry miR-21a-5p exacerbate IBD lesions by downregulating the E-cadherin expression and innate lymphoid cell activation in enterocytes [53].
Rather limited research exists on the role of SEs in IBD [47]. In the one and only study that has been reported to date, Zheng et al. extracted SEs from IBD patients and healthy individuals, and more than 2000 proteins were identified using LC-MS [47]. In total, eight of these proteins, which were related to inflammation and proteasome activity, were selected for further analysis [47]. The expression of proteasome subunit alpha type 7 (PSMA7) was significantly elevated in the IBD patients, relative to the healthy controls [47]. PSMA7 is involved in the proteasome activity and inflammatory responses in IBD, and thus can potentially serve as a crucial biomarker for IBD screening [47].

2.2. Neurodegenerative Disease

Exosomes from multiple sources have been extensively studied in neurodegenerative diseases [22,48]. In this section, we will discuss Alzheimer’s and Parkinson’s disease, their pathophysiology and ocular manifestations, and the advances in the applications of exosomes [109,110,111], as shown in Table 2.

2.2.1. Alzheimer’s Disease

Alzheimer’s disease (AD) is a neurodegenerative disease that is characterized by progressive cognitive impairment and dementia [112,113,114,115]. The neuropathological lesions in the AD brain include the extracellular deposition of amyloid-beta (Aβ) protein, forming senile plaques, and the hyperphosphorylated tau (p-Tau) protein, forming neurofibrillary tangles [22,112,116,117,118]. The Aβ proteins that are found in the senile plaques of AD are also major constituents of the retinal drusen characteristic of age-macular degeneration, suggesting that AD and AMD share pathogenic mechanisms [119,120]. A significant increase in drusen has been reported in the retinas of AD patients compared to those of healthy patients [121,122]. Multiple studies have also demonstrated a decreased retinal nerve fiber layer thickness in AD patients [123,124,125,126], a decreased contrast sensitivity [127,128,129], and slower, hypometric saccades [130,131,132], when compared to their healthy counterparts. Recently, the hallmark biomarkers of AD, Aβ, and p-Tau have been detected in the retinas of mice models, and methods for the in vivo imaging of these retinal biomarkers require further validation [133,134,135].
It has been shown that exosomes transport Aβ and tau protein, facilitating the propagation of the Aβ and tau pathologies [118,136,137]. Saman et al. observed that neuronal exosomes mediate the secretion of the pathologic tau protein from neurons, reporting tau overexpression in the neuroblastoma cells that recruit mitochondrial proteins, disrupting neural synapses and axons [138]. Higher levels of Aβ and p-Tau proteins have also been detected in the serum exosomes of AD patients, compared to those of healthy controls [139,140]. Goetzl et al. found that, compared to those isolated from the controls, the astrocyte-derived exosomes from AD patients contained significantly higher levels of β-site amyloid precursor protein-cleaving enzyme 1 (BACE-1) and amyloid precursor protein (APP) [137]. Since BACE-1 is required to cleave APP into Aβ peptides [141,142], the astrocyte-derived exosomes that carry BACE-1 represent critical tools for testing the AD therapies that inhibit BACE-1 [137].
Astrocyte-derived exosomes express elevated levels of complement C3 proteins in patients with mild cognitive impairment (MCI) progressing to AD dementia, compared to patients with MCI that remained stable over 3 years [136]. Interestingly, Chiarini et al. showed that exosomes mediate the release of p-Tau from astrocytes, and the levels of p-Tau increased after exposure to Aβ25-35 [143]. Sun et al. studied the urinary exosomes in AD patients for the first time, and reported significantly higher levels of Aβ1-42 and P-S396-tau in the AD patients [144]. Notably, neuronal exosomes were isolated from the induced pluripotent stem cells of a familial AD patient with an A246E mutation to presenilin-1, and injected into the hippocampi of wild-type C57BL/6 mice [145]. Th mice showed increased tau inclusions and an accelerated tau phosphorylation [145]. This study revealed exosomes as a potential mediator of the tau dysregulation that is associated with familial AD [145].
There have been several reports of neuronal exosomes slowing AD progression [109,146]. Yuyama et al. demonstrated the possible neuroprotective roles of neuronal exosomes by facilitating the uptake and clearance of Aβ with microglial cells [147]. The blockade of phosphatidylserine, a surface protein on exosomes, inhibited the exosome-mediated uptake of Aβ into microglia [147]. The inhibition of neutral sphingomyelinase 2 activity reduced the exosome secretion, while the knockdown of sphingomyelin synthase 2 increased the exosome secretion [147]. In addition, the exosomes that are derived from human cerebrospinal fluid (CSF) can sequester Aβ oligomers via surface proteins, such as cellular prion protein [146,148]. Recently, it has been shown that the exosomes from human neural stem cells reduce the Aβ plaque accumulation and microglial activation, protecting against synaptic atrophy [109]. Thus, neuronal exosomes have the potential to mitigate the Aβ-mediated impairment of synaptic plasticity and long-term potentiation [109].
The effects of MSC-derived exosomes on the alleviation of the neuroinflammation in AD is also well described [149,150,151]. MSCs have been observed to slow the memory deterioration of AD mice by increasing anti-inflammatory cytokines, including IL-10, and decreasing the pro-inflammatory cytokines, IL-1β and TNF-α [152]. Exosomes that were isolated from human umbilical cord MSCs were injected into AD mice and found to promote the clearance of Aβ plaques and reduce the levels of pro-inflammatory cytokines [149,153]. Wang et al. showed that exosomes from bone marrow MSCs decreased the Aβ deposition by activating the sphingosine kinase/spingosine-1-phosphate signaling pathway [150], and at the same time, improved spatial learning and cognitive function and reduced the levels of the BACE1, Aβ1-40, and Aβ1-42 peptides [150]. Since BACE1 cleaves the amyloid precursor protein to form the Aβ1-40 and Aβ1-42 peptides, reducing the BACE1 levels is critical to slowing the Aβ accumulation and disease progression [142,154]. Using a neural cell culture model that simulated AD by overexpressing APP, Chen et al., demonstrated that mesenchymal stem cell-derived exosomes reduced Aβ production [151]. Exosomal miR-29a enhanced the expression of the genes that are involved in synaptic plasticity, by downregulating the expression of histone deacetylase 4, which is markedly increased in AD patients [151,155].
While studies on the exosomes in AD have been plenty, SEs specifically have only recently been examined [22]. Rani et al. used a nanoparticle tracking analysis to quantify and analyze the SEs in 10 cognitively impaired patients, 5 AD patients, and 12 healthy controls [22]. The total concentration of the SEs was significantly higher in the cognitively impaired and AD patients, when compared to the healthy controls [22]. Furthermore, the elevated levels of SEs were correlated with a decrease in the ACEIII value, indicating an increased cognitive impairment and disease severity [22]. These results confirmed that the oligomeric forms of the Aβ protein and p-Tau protein were significantly higher in the cognitively impaired and Alzheimer’s disease patients [22]. Together, these findings demonstrate the utility of a nanoparticle tracking analysis to quantify the SEs’ concentration, and to serve as a cost-efficient screening method for the early detection of AD [22].

2.2.2. Parkinson’s Disease

Parkinson’s disease (PD) is a movement disorder that occurs due to the degeneration of the pigmented neurons in the substantia nigra, involving Lewy bodies that contain α-synuclein protein [48,156,157,158]. Its hallmark clinical features include bradykinesia (slowness of movement), resting tremors, postural disequilibrium, and ataxia [158,159]. The bradykinesia manifests not only in the limbs, but also in the eyes, as impaired smooth pursuit [160,161]. Other oculomotor abnormalities in PD include an impaired convergence [162,163,164,165], diplopia [165,166,167], and an impaired vertical gaze [160,168]. PD has also been associated with a higher incidence of dry eye [164,169,170,171] and glaucoma [172,173].
PD-derived exosomes can trigger the formation of α-synuclein oligomers, which are then packaged in exosomes for disease propagation [174]. In addition, evidence of α-synuclein transport via the exosomes from CSF to the venous circulation implicates the role of these exosomes in PD progression [175,176]. Stuendl et al. showed that the exosomes from the CSF of PD patients carried higher levels of α-synuclein, and suggested that CSF exosomes may disseminate the PD pathology by triggering the oligomerization of α-synuclein [177]. Similarly, treatment with human α-synuclein preformed fibrils induced the microglia to release exosomes containing α-synuclein [178]. By degrading the lysosomal structural protein LAMP2, the α-synuclein preformed fibrils also impaired the lysosomal breakdown of exosomal α-synuclein from microglia, indicating that microglial-derived exosomes contribute to the α-synuclein aggregation in neurons [178,179,180]. Recently, Si et al., reported that the exosome secretion by microglial cells is increased by the NOD-like receptor family pyrin domain, containing three protein (NLRP3) inflammasome, which is known to be involved in the neuroinflammation of PD [181].
Elevated α-synuclein levels have also been found in plasma neural-derived exosomes [182,183]. After the plasma exosomes from PD patients were injected into mice brains, the microglia showed an increased uptake of these exosomes and the release of exosomal α-synuclein, implying that microglia may facilitate the transmission of exosomal α-synuclein [184]. The plasma exosomes were also found to dysregulate the autophagy in mouse microglia, as measured by the increased accumulation of α-synuclein in the intracellular and extracellular spaces [111,184]. Besides α-synuclein, serum exosomes have been shown to carry other biomarkers of PD [185,186]. For example, miR-137, which is known to induce the oxidative stress of neurons in PD [186], was detected in increased levels in the serum exosomes of PD patients, and was shown to downregulate oxidation resistance 1 (OXR1) [185]. The depletion of miR-137 or the upregulation of OXR1, on the other hand, alleviated PD-induced oxidative stress [185].
The neuroprotective effects of exosomes in PDs have also been reported [110,187,188]. The treatment of PD mice with blood-derived exosomes from healthy human subjects was found to reduce the loss of dopaminergic neurons, alleviate oxidative stress and inflammation, and improve the motor coordination in PD mice [188]. In addition, this exosome treatment significantly reduced the mRNA expression of the pro-inflammatory cytokines, IL-1beta, IL-6, and TNF- a, and increased the mRNA levels of the anti-inflammatory cytokines IL-4, IL-10, and TGFB, indicating that exosomes can attenuate neuroinflammation [188]. The exosomes secreted by human umbilical cord MSCs demonstrated a neuroprotective role in PDs [110], by crossing the blood–brain barrier to reduce the apoptosis of the dopaminergic neurons in the substantia nigra [110]. Specifically, miR-188-3p, which is carried by MSC exosomes, suppressed neuron loss by targeting the NLRP3 autophagy pathway [187]. The treatment with MSC-derived exosomes also promoted the proliferation of neuronal cells that were subjected to 6-hydroxydopamine (6-OHDA)-mediated oxidative stress [110]. These findings illustrate the protective ability of MSC exosomes in PD.
Although limited, there is evidence for SEs having a role in PD propagation [48,189]. In a study of 74 PD patients and 60 healthy controls, Cao et al. reported significantly higher levels of oligomeric α-synuclein and higher ratios of oligomeric α-synuclein to the total α-synuclein in the SEs from PD patients, relative to the healthy controls [48]. This was supported by another study of 18 PD patients and 15 healthy controls that found the ratio of hyperphosphorylated α-synuclein to the total α-synuclein to be significantly higher in the PD-derived SEs [189]. These data support the idea that the pathogenic mechanism of PD involves SEs disseminating the misfolded α-synuclein proteins in the brain [48,189].

2.3. Malignant Neoplasms

Exosomes have been used to identify the markers for tumor migration, proliferation, and metastasis [190,191]. It has been found that exosomes contain miRNA and circulating tumor genes (ctDNA), which are both involved in intercellular communication in the tumor microenvironment [192,193,194]. In this section, we will discuss various cancers, their ocular effects, and the roles of exosomes in cancer development and progression. A summary of the cancers, exosomal biomarkers, and their effects, are shown in Table 2.

2.3.1. Oral Cancers

Oral cancers, also known as head and neck cancers, are malignancies that often occur in the oral cavity, especially the tongue and the floor of the mouth, the oropharynx, and the esophagus [195,196,197,198]. Oral squamous cell carcinoma (OSCC) is the most common type of oral cancer in the world [199]. The risk factors that are involved in the development of oral cancer in Western countries include the male sex and an age of over 65 years [195,197,199]. Its etiologic agents include smoking and alcohol consumption [195,199,200]. While the risk factors and etiology of oral cancers have been extensively studied, little is known about the ocular effects of oral cancers [201,202,203]. In one of the few studies that have been reported, an increased incidence of dry eye disease, as indicated by a reduced tear film break up time and a decreased tear production, has been found in oral cancer patients [201]. Few cases of the ocular metastases of oral cancer have been reported, occurring through choroidal spread [202,203].
Although limited, the findings on exosomes in oral cancers have demonstrated that they can both inhibit and promote cancer cell growth [49,50,204]. Human bone marrow MSC-derived exosomes that overexpress miR-101-3p inhibit oral cancer progression by downregulating COL10A1 [204]. However, plasma-derived exosomes showed immunosuppressive effects that facilitate the progression of head and neck squamous cell carcinomas (HNSCC) and esophageal cancer [50]. For example, miR-93-5p and miR-19b-3p, transferred by plasma exosomes, promote the proliferation of esophageal cancer cells by inhibiting PTEN expression [49,205]. Furthermore, exosomes from the plasma of HNSCC patients with active disease, compared to the plasma exosomes from patients with no evident disease, more effectively inhibited CD4(+) T-cell proliferation, promoted the apoptosis of CD8(+) T cells, and increased the regulatory T-cell suppressor functions [206]. Notably, PD-L1, a known tumorigenic factor that is carried by the plasma exosomes from HNSCC patients, is linked to disease progression, as the blocking of PD-L1 signaling attenuates immune suppression [50].
The literature on SEs in relation to oral cancers is more prolific [207,208,209]. Wang et al. investigated the potential of human papillomavirus DNA that was extracted from human saliva as a biomarker for HNSCC. The authors observed that the tumor DNA that was detected in all the early stage disease patients (n = 10), and in 95% of the late-stage disease patients (n = 37), was significantly higher than the proportions of the tumor DNA that was found in the plasma of the HNSCC patients [208]. To examine the differences in the exosomal miRNA of HNSCC patients, relative to the controls, Langevin et al. cultured four discrete HNSCC cell lines, originating from four different sites in the upper digestive tract: H413 (buccal mucosa), Detroit 562 (pharynx), FaDu (hypopharynx), and Cal 27 (tongue), as well as human gingival epithelial cells from healthy donors for comparison [207]. Higher levels of miR-486-5p, miR-486-3p, and miR-10b-5p were detected in the HNSCC patients relative to the controls [207]. Importantly, miR-486-5p was also found in the early-stage lesions, implicating its role in early disease detection [207]. Furthermore, miR-302b-3p and miR-517b-3p were expressed in the SEs from oral squamous cell carcinoma (OSCC) patients, and miR-512-3p and miR-412-3p were higher in the OSCC patients [209]. Elevated levels of GOLM1-NAA35 chimeric RNA were reported in esophageal squamous cell carcinoma tissues [210]. Other studies on OSCC patients and healthy controls demonstrated that miR-29a-3p promoted tumor growth by inducing M2 subtype macrophage polarization via the SOCS1/STAT6 signaling pathway [211]. An increased expression of miR-29a-3p [211] and miR-31 [212] was found in the exosomes of OSCC patients. These exosomal miRNA levels were significantly decreased after a tumor resection, indicating their utility in monitoring cancer progression [212].

2.3.2. Breast Cancer

Breast cancer is the most common type of cancer affecting women worldwide [213,214,215]. Although rare, the ocular metastases of breast cancer have been reported in both female and male patients [216]. The breast is a common origin site of ocular metastatic tumors, accounting for 49% of patients [216]. Since most ocular metastases occur through hematogenous dissemination, the uveal tract, especially the choroid, due to its vascularity, is the most common site of origin for breast cancer metastases [217,218]. A common complication of metastatic choroidal disease in breast cancer patients is vision deterioration, due to a macular invasion of the tumor or fluid accumulation in the fovea [219,220].
Exosome studies of breast cancer have gained recent traction [221,222]. Sun et al. demonstrated that RAB22A, a proto-oncogene involved in the production, trafficking, and metabolism of exosomes, upregulates exosome-mediated breast cancer cell proliferation, invasion, and migration [223]. However, miR-193b downregulates the oncogenic effects of RAB22A, hindering exosome-induced cancer cell metastasis [223]. Additionally, oncogenic miRNAs, miR-21 and miR-200c, have been detected in higher levels in the tear exosomes of breast cancer patients compared to healthy subjects [224]. Notably, miR-21 was increased by nearly 3-fold and miR-200c was increased by 15-fold, suggesting that these tear exosomes can be a useful source of diagnostic and prognostic markers for breast cancer [224]. Ando et al. found that the matrix metalloproteinase-1 (MMP-1) in the urinary exosomes from breast cancer patients was significantly elevated compared to the healthy controls [225]. In contrast, the miR-21 expression in the patients was significantly lower than in the controls, indicating that MMP-1 and miR-21 may be potential screening markers for breast cancer [225]. Shtam et al. reported that the plasma exosomes from healthy donors facilitated the migration and transwell invasion of breast cancer cells, and promoted their metastatic spread [221]. This effect was mediated by the interactions of exosome surface proteins with breast cancer cells, stimulating focal adhesion kinase signaling in the breast cancer cells [221]. Shtam et al. concluded that plasma exosomes have the potential to trigger the metastasis of breast cancer cells [221].
While other types of exosomes, including those from plasma, tears, and urine, have been shown to induce breast cancer progression, some studies have demonstrated the therapeutic effects of MSCs in breast cancer [222,226,227,228,229]. MSC-derived exosomes inhibit the VEGF expression and angiogenesis of breast cancer cells, by transporting miRNA-100 via the mTOR/hypoxia-inducible factor (HIF)-1-α signaling pathway [226,227]. Furthermore, the human umbilical cord MSC-derived exosomes that carry miR-148b-3p inhibited the breast cancer cell proliferation, migration, and invasion [228]. Recently, exosomes from adipose MSCs inhibited breast cancer metastasis and epithelial-to-mesenchymal transition (EMT) via the downregulation of DNA repair genes (PARP1 and CCND2) and cancer stem cell surface markers (CD44 and ALDH1) [229]. Similarly, these adipose MSC-derived exosomes, loaded with miR-381, significantly reduced the expression of EMT-related genes, inhibiting the proliferation, migration, and invasion of breast cancer cells [222].
There have been several studies of SEs in breast cancer [230,231]. Zhang et al. reported higher levels of carbonic anhydrase 6 (CA6) and insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) in the saliva of breast cancer patients compared to controls [232]. Notably, elevated levels of CA6 and CA15-3 have been detected in the saliva of breast cancer patients [233,234]. Similarly, TCTP1 and IGF2BP1 promote the proliferation of various cancers, including breast cancer, and TCTP1 overexpression stimulates the degradation of the tumor suppressor gene, p53 [230,235]. Lau et al. treated salivary gland exosomes with breast-cancer-derived exosomes to examine the interactions between them [231]. The authors detected 88 proteins in the treated salivary gland exosomes at levels that were 1.5 times higher than those in the control group, and 66 mRNAs that were differentially expressed by the treated exosomes, indicating the ability of breast-cancer-derived exosomes to modify the content of both the proteins and mRNAs in SEs [231]. The breast-cancer-derived exosomes also upregulated the total RNA that was expressed by the salivary gland exosomes [231]. This increase in the total RNA was significantly decreased after the salivary glands were exposed to the transcription inhibitor actinomycin D [231]. These results suggest that the upregulated RNA transcription that was detected in the SEs was mediated by the interplay between the salivary gland exosomes and breast-cancer-derived exosomes [231].

2.3.3. Colorectal Cancer

Colorectal cancer is the third most common cancer in men and second most common cancer in women worldwide [213]. Colorectal cancer is the second cause of cancer-related deaths in Western countries, accounting for 9% of all cancer-related deaths [236]. While rare, several cases of intraocular metastasis from colorectal cancer exist, disseminating via the choroid [237,238].
Although scarce, reports on the exosomal markers for colorectal cancer exist [239,240,241]. Xiao et al. reported that cytokeratin 19 (CK19) was enriched in the exosomes that were derived from colorectal cancer cells, that carbohydrate antigen 125 (CA125) was elevated in the exosomes from metastatic colorectal cancer cells, and that tumor-associated glycoprotein 72 (TAG72) was detected in the exosomes from 5-fluorouracil (5-FU)-resistant colorectal cancer cells [240]. CK19, CA125, and TAG72 were also found in the interstitial-fluid-derived exosomes and serum-derived exosomes of colorectal cancer patients [240]. Additionally, Li et al. found a higher concentration of circular RNAs (circRNAs) that were secreted by the serum exosomes from colorectal cancer patients, supporting the utility of circRNAs in cancer diagnosis [241]. Recently, angiopoietin-like protein 1 (ANGPTL1), which was detected in exosomes that were derived from human colorectal cancer cells, has been shown to inhibit the liver metastasis of colorectal cancer cells by blocking MMP9-induced vascular leakiness via the JAK2-STAT3 pathway [239].
The SEs in colorectal cancer have attracted recent interest, but much is still unknown about their function in its pathogenesis [242,243]. Sazanov et al. detected, using a qRT-PCR, a significantly higher miR-21 expression in the SEs from colorectal cancer patients compared to controls, and this miR-21 expression demonstrated a high diagnostic sensitivity and specificity (97% and 91%, respectively) [242]. Other salivary miRNAs, including miR-186-5p, miR-29a-3p, miR-29c-3p, miR-766-3p, and miR-491-5p, were significantly elevated in colorectal cancer patients [243]. Together, the literature suggests that SEs may not only represent a critical tumor screening tool, but may also be used to treat malignant tumors, by impeding tumor invasion and growth [242,243].

2.3.4. Lung Cancer

Lung cancer is the leading cause of cancer mortality in men and women in the United States, accounting for nearly 25% of cancer deaths [213]. Smoking is the number one risk factor for developing and dying from lung cancer [213]. Like most ocular metastases from solid tumors, orbital involvement from primary lung cancer most commonly affects the choroid [244,245,246]. The prognosis of lung cancer with choroidal metastasis is poor, with a median survival of 6-13 months [247]. There have also been a few rare cases of iris metastases, secondary to lung tumors [248,249,250].
Exosomal applications in lung cancer have gained attention in recent years. Rahman et al. observed that exosomes from metastatic lung cancer cells induced vimentin expression and EMT, as well as migration, invasion, and proliferation in non-cancerous human bronchial epithelial cells [251]. The authors concluded that lung-cancer-derived exosomes could drive healthy recipient cells towards EMT [251]. Xiao et al. found that the exosomes secreted by A549 lung cancer cells decreased the sensitivity of the A549 cells to cisplatin, and the increase in chemoresistance may be mediated by the exosomal transport of miRNAs [252]. More recently, exosomes from lung cancer bronchoalveolar lavage fluid were found to promote the migration and invasion of A549 lung cancer cells by carrying E-cadherin [253]. Macrophage-derived exosomes have also been shown to promote cisplatin resistance in lung cancer [254]. By delivering miR-3679-5p to A549 lung cancer cells, macrophage-derived exosomes enhance aerobic glycolysis and chemoresistance, via the NEDD4L/c-Myc signaling cascade [254].
Table 2. Exosomes in non-ocular diseases.
Table 2. Exosomes in non-ocular diseases.
DiseaseExosome TypeBiomarkersEffectReference
Alzheimer’s DiseaseSalivaAβ, p-Tau
Aβ1-42, P-S396-tau
Disease propagation via Aβ and tau deposition [14]
Blood[131,132]
Urine[136]
AstrocytesBACE-1, sAPPβ,
Complement C3,
p-Tau
[129]
[128]
[135]
CSFCellular prion proteinNeuroprotective; reduce exosome uptake of Aβ[140]
MSCsBACE1, Aβ1-40, Aβ1-42Inhibit Aβ deposition[142]
miR-29aIncrease synaptic plasticity via HDAC4 downregulation[143,147]
Parkinson’s DiseaseSalivaα-synuclein oligomersDisease propagation via α-synuclein oligomerization[40,181]
CSF[169]
MicrogliaInhibit lysosomal breakdown of α-synuclein [170]
SerumUptake of α-synuclein into microglia[176]
miR-137Increase oxidative stress via OXR1 inhibition[177]
Oral Lichen PlanusSalivamiR-4484, miR-146a, miR-155Unknown[37,38]
T cellsMIP-1 alpha/betaT-cell migration; apoptosis of keratinocytes [55,56]
PeriodontitisSalivamiR-140-5p, miR-146a-5p, miR-628-5p
LPS, 5mC methylation
Unknown[43,68]
Periodontal ligament stem cellsmiR-17-5pInhibit inflammation and angiogenesis[65]
Sjorgen’s SyndromeSalivamiR-768-3p, miR-574-3pUnknown[77]
Anti-Ro/SSA, anti-La/SSB, Sm ribonucleoproteinsAutoantigen presentation to lymphocytes[79]
CD44 antigen; NGALT cell activation; neutrophil activation [80]
T cellsmiR-142-3pDecrease protein production from salivary glands via cAMP inhibition[73]
Inflammatory Bowel DiseaseSalivaPSMA7Proteasome activity and inflammatory response[39]
MSCsmiR-326Inhibit ubiquitination via NF-kappaB downregulation[98]
T regulatory cellsmiR-195a-3pReduce inflammation[100]
MacrophagesmiR-21a-5pExacerbate IBD via E-cadherin inhibition[45]
Oral squamous cell carcinomaSalivamiR-302b-3p, miR-517b-3p, miR-512-3p, miR-412-3pUnknown[201]
miR-31, miR-29a-3pPromote M2 subtype macrophage polarization[203,204]
MSCsmiR-101-3pInhibit cancer progression via COL10A1 downregulation[196]
Esophageal CancerSalivaGOLM1-NAA35 chimeric RNA Unknown[202]
PlasmamiR-93-5p; miR-19b-3pProliferation of esophageal cancer cells via PTEN inhibition[41,197]
Head and NeckSalivaHuman papillomavirus DNA,
miR-486-5p, miR-486-3p, miR-10b-5p
Unknown[199,200]
Breast cancerSalivaCA6, CSTA, TPT1, IGF2BP1Unknown [224]
UrineMMP-1Pro-angiogenic[217]
MSCsmiRNA-100,
miR-148b-3p,
miR-381
Inhibit angiogenesis and cancer cell proliferation[214,218,220]
Colorectal cancerSalivaANGPTL1Blocks metastasis via MMP9 inhibition[238]
miR-21,
miR-186-5p, miR-29a-3p, miR-29c-3p, miR-766-3p, miR-491-5p
Unknown[241,242]
Lung cancerSalivaBPIFA1, CRNN, MuC5B, IQGAPUnknown[254]
Lung cancer bronchoalveolar lavage fluidE-cadherinPromote cancer cell migration and invasion[252]
MacrophagemiR-3679-5pPromote aerobic glycolysis and chemoresistance[253]
The literature on using SEs and lung cancer mechanisms is limited. In one of the few studies that have been reported, Sun et al. compared the protein composition of the SEs in lung cancer patients with that of normal subjects, and verified the presence of four proteins that are specific to lung cancer tissue (BPIFA1, CRNN, MuC5B, and IQGAP) [255].

3. Exosomes in Ocular Diseases

In addition to non-ocular diseases, exosomes have been implicated in the development and/or progression of several ocular diseases [256,257,258,259]. The subsequent sections will highlight the roles of exosomes in diabetic retinopathy, corneal disease, age-related macular degeneration, uveal disease, and glaucoma. Table 3 summarizes the ocular diseases, exosome biomarkers, and their effects.

3.1. Diabetic Retinopathy

Diabetic retinopathy (DR) is a complication of diabetes that is caused by damage to the blood vessels in the retina [260,261]. DR remains the leading cause of blindness and visual impairment in working-aged adults and is a growing public health problem [261,262]. The global diabetes prevalence in 2021 was estimated to be 537 million (10.5%), and this figure is projected to increase to 783 million by 2040 (12.2%) [263].
Non-proliferative DR is the early stage of the disease, in which the blood vessels in the retina are weakened, leading to the formation of microaneurysms and the swelling of the macula [260,264]. The early histological features of DR include retinal endothelial cell loss and the thickening of the retinal vascular basement membrane, correlating with increased fibronectin levels [265,266,267]. Proliferative DR is the more advanced form of the disease, marked by retinal hypoxia and resulting in neovascularization, in which new, fragile blood vessels begin to grow in the retina and into the vitreous [268]. If left untreated, PDR can cause retinal detachment, severe vision loss, and blindness [268,269,270,271]. Since the pathogenesis of proliferative DR has been shown to involve the neovascularization of retinal cells, exosomes contribute to the disease development by regulating the angiogenic factors [272,273]. For example, Tokarz et al. found that, relative to the exosomes of healthy controls, the exosomes of diabetic patients carried significantly higher pro-inflammatory and pro-angiogenic factors, such as basic fibroblast growth factor, TNF-α, VEGF receptor 2, and angiopoietin-2 [273]. More recently, Maisto et al. exposed retinal photoreceptors to high glucose concentrations (30 mM) and found a significant upregulation of VEGF and downregulation of anti-angiogenic miRNAs, including miR-20a-3p, miR-20a-5p, and miR-20b, in the exosomes that were released by the retinal cells [272].
Plasma exosomes have been shown to carry critical biomarkers and induce oxidative damage in DR [274,275,276]. Specifically, plasma exosomes carry elevated amounts of peroxisome proliferator-activated receptor gamma (PPAR-γ) in the aqueous humor and vitreous fluid of proliferative DR patients, relative to healthy controls [277]. A significant positive correlation between the PPAR-γ and VEGF concentrations was also found, together with notable increase in the PPAR-γ levels, which was correlated with the clinical progression of DR, delineating the role of PPAR-γ in the pathogenesis of DR [277]. Huang et al. discovered that plasma exosomes also contain IgG, which activates the classical complement pathway and inflammatory responses, and showed that these exosomes are increased in DR [278]. Huang et al. later found that IgG-carrying exosomes cause damage to the retinal endothelial cells by initiating membrane attack complex (MAC) formation and activating the complement system [274]. Zhang et al. similarly noted that exosomes from platelet-rich plasma (PRP-Exos) exacerbated hyperglycemia-induced retinal ischemia [275]. PRP-Exos were upregulated in diabetic rats via the TLR4 signaling pathway, mediating the inflammation of the retinal endothelial cells [275].
Multiple studies have implicated the therapeutic potential of exosomes in diabetic retinopathy [279,280,281]. Using an oxygen-induced retinopathy mouse model, Moisseiev et al. investigated the effect of exosomes on retinal ischemia, by intravitreally administering exosomes that were derived from human MSCs [282]. Retinal thinning and angiogenesis were significantly reduced in the ischemic retinas that were treated with the MSC-derived exosomes compared to the eyes that were treated with saline [282]. A similar therapeutic role of these MSC-derived exosomes was exhibited in a rabbit model of DR [281]. The administration of the MSC-derived exosomes from rabbit adipose tissue caused the regeneration of the normal retinal layers and increased the expression of miRNA-222, indicating that exosomes may induce retinal repair via the miRNA-222 transfer to damaged cells [281].
Like MSC-derived exosomes, exosomes from retinal cells have also been implicated in the retinal repair of DR lesions [279,280,283]. For instance, Liu et al. demonstrated that exosomes carry circular RNA-cPWWP2A, which indirectly modulates endothelial cell activity via the inhibition of miR-579 [283]. The upregulation of cPWWP2A expression, due to diabetes-induced stress, was shown to reduce retinal endothelial damage [283]. Another study, a year later, found that the retinal exosome depletion by GW4869 impeded the transport of photoreceptor-derived miR-124-3p to the inner retina [279]. Exosomal inhibition by GW4869 also aggravated the retinal lesions and induced further inflammation and damage to the photoreceptors, implying a role of retinal exosomes in protecting against retinal degeneration [279]. Interestingly, Gu et al. suggested that retinal pigment epithelium (RPE)-derived exosomes suppress the pathologic fibrosis in proliferative DR by transferring miR-202-5p to human umbilical vein endothelial cells (HUVECs). miR-202-5p, which is carried by exosomes, negatively regulated TGF-β2, resulting in the inhibition of the HUVEC proliferation and tube formation [280].

3.2. Retinitis Pigmentosa

Retinitis pigmentosa is a retinal degenerative disease, in which rod photoreceptors are lost due to inherited gene mutations [284,285]. While this loss of rods leads to night blindness, the progression of the disease may cause total vision loss when the cone photoreceptors are irreversibly damaged [284]. Previous studies have demonstrated that poly-ADP-ribose polymerase (PARP) hyperactivity may be involved in the retinal degenerative process of retinitis pigmentosa [286,287]. Vidal-Gil et al. examined the relationship between PARP inhibition and exosome secretion [288]. The authors found that CD9, a tetraspanin protein on the surface of exosomes, was in the outer nuclear layer (ONL) and inner retina, suggesting that a significant proportion of exosomes may be secreted from retinal photoreceptors [288]. Furthermore, after the inhibition of the PARP activity, the CD9 expression was reduced in the ONL and inner layers of the retina, while the photoreceptor degeneration was significantly improved [288]. Thus, decreasing the secretion of CD9-expressing retinal exosomes might help to alleviate photoreceptor damage [288].

3.3. Age-Related Macular Degeneration

Age-related macular degeneration (AMD) is a leading cause of central vision loss among the elderly [289]. The pathogenesis of the early, non-exudative, “dry” type of AMD involves the formation of drusen, a lipid-like material that deposits on Bruch’s membrane in the macula of the retina [290,291,292,293]. These drusen deposits disrupt the retinal blood supply, resulting in a loss of photoreceptors and vision loss [291,292,293,294]. The late, exudative, “wet” type of AMD is characterized by macular neovascularization and retinal endothelial leakage, leading to severe, rapid vision loss [291,293,295,296].
Mutations in complement factor H and the dysfunction of complement protein C3 are associated with an increased risk for the development of AMD [297,298]. Wang et al. observed that exosomes released by the RPE of AMD patients are coated with C3 and can interact with complement factor H (CFH), and proposed that the mutated CFH in AMD may disrupt the exosomal release of proteins and the subsequent incorporation of these proteins into drusen [290]. The proteins CD63, CD81, and LAMP2 were also detected in the RPE-derived exosomes and drusen of AMD patients, but not in the healthy controls, providing evidence that RPE-derived exosomes contain proteins that promote drusen production [290]. Kang et al. identified the exosomal proteins secreted by RPE that may serve as potential biomarkers for AMD [299]. Specifically, higher levels of cathepsin D and cytokeratins 8 and 14 were found in the aqueous-humor-derived exosomes of AMD patients [299]. The authors concluded that the upregulation of these proteins may be due to a reactive response against the oxidative stress mediated by the autophagy-lysosomal pathway [299].
Several studies have suggested that the macular neovascularization and retinal endothelial leakage of exudative AMD may be mediated by the RPE-derived exosomes that express VEGF receptors [300,301]. A recent in vitro assay found that the serum-derived exosomes from AMD patients were upregulated in miR-19a, miR-126, and miR-410, and formed significantly more endothelial tubules, relative to the serum-derived exosomes from controls [302]. Bioinformatics analyses confirmed that these exosomal miRNAs were associated with the VEGF/angiogenesis and apoptosis signaling pathways [302]. These findings demonstrated a pathogenic role of serum exosomes in AMD by promoting the neovascularization and apoptosis of retinal cells [302]. miR-126, miR-410, and miR-19a have been previously implicated in retinal disease [303,304,305]. miR-126 promotes VEGF expression and retinal neovascularization, while miR-410 suppresses VEGF-mediated neovascularization [303,304,305]. Interestingly, increasing the miR-19a levels in RGCs has been shown to promote axon regeneration in vivo after an optic nerve crush in mice, and in RGCs from human donors [306].
While the exosomes from serum and RPE exhibit pro-angiogenic effects, the exosomes that are released from retinal astroglial cells (RACs) have anti-angiogenic properties [307]. In a model of laser-induced choroidal neovascularization (CNV), RAC-derived exosomes inhibited CNV and vascular lesions, while the exosomes that were secreted by RPE did not [307]. Anti-angiogenic agents, such as endostatin and MMP3, were detected in the RAC-derived exosomes [307]. These findings indicated that the exosomes that are released from RACs downregulate angiogenesis by suppressing the migration of macrophages and vascular endothelial cells, both of which have been associated with the development of exudative AMD [307]. More recently, it has been reported that exosomes that are released from mouse neural progenitor cells (NPCs) have therapeutic effects in AMD, by slowing retinal degeneration [256], exhibiting inhibitory effects on microglial cells, and mitigating photoreceptor apoptosis and the atrophy of the ONL [256].

3.4. Corneal Diseases

The role of exosomes in corneal wound healing has been examined in various studies [308,309,310,311]. Corneal fibroblasts have been shown to secrete exosomes that deliver MMP14 and other angiogenic proteins to endothelial cells, and MMP14 is involved in the incorporation of MMP2 into corneal fibroblast exosomes [311]. These findings suggest that the MMP14 in exosomes may be a key therapeutic target for angiogenesis and the mediation of the cell–cell communication in the cornea [311]. The same group of investigators also found that the exosomes that are secreted by mouse corneal epithelial cells, upon fusing to target keratocytes, stimulated the differentiation of myofibroblasts and the expression of alpha-smooth muscle actin (α-SMA), a known marker of fibrosis [310]. As myofibroblast differentiation is implicated in corneal wound closure and neovascularization, corneal epithelial-derived exosomes may be important therapeutic targets for corneal disease [310]. Leszczynska et al. found that exosomes from limbal stromal cells (LSCs) accelerated the limbal epithelial stem cell (LESC) proliferation and wound healing via Akt phosphorylation and the upregulation of LESC markers, including keratin 15 [312].
More recently, McKay et al. found that exosomes were released by human corneal epithelial cells, fibroblasts, and endothelial cells (Figure 4 and Figure 5), indicating the role of exosomes in the cell–cell signaling amongst these cell types [313]. The authors also reported the expression of α-SMA (Figure 6), fibronectin (Figure 7 and Figure 8), thrombospondin-1 (TSP-1) (Figure 9), and by human corneal epithelial cells, when co-cultured with corneal stromal fibroblasts [313]. This increased α-SMA expression and enhanced contractility suggest that epithelial exosomes promote stromal cell differentiation into myofibroblasts, which mediates wound closure [313]. The expression of the provisional matrix proteins, fibronectin and TSP-1, indicates that exosomes may also contribute to basement membrane regeneration and scar formation [313].
Wound healing properties of the exosomes from corneal stem cells have also been reported [308,309,314]. Samaeekia et al. investigated the effect of these exosomes on wound healing in vitro using a scratch assay, and in vivo using epithelial debridement wounds in mice [308]. They found that the exosomes that were derived from human corneal MSCs accelerated the corneal epithelial wound healing, both in vitro and in vivo, demonstrating their therapeutic potential in corneal epithelial wound repair [308]. Another study using a mouse model illustrated the therapeutic effects of MSC-derived exosomes on corneal wound healing [309]. Exosomes, via the transfer of miRNAs to corneal tissue, inhibited neutrophil invasion into the wounded area and promoted the regeneration of normal collagen in the corneal tissue [309]. These exosomes also attenuated the fibrotic scarring of the stroma, as measured by the reduced expression of the fibrotic genes encoding for collagen III and α-SMA [309]. When the packaging of miR159a into the exosomes was blocked by knocking out Alix protein, the exosomes with reduced miRNA were less effective at suppressing corneal scarring and promoting tissue regeneration [309]. These results demonstrate that corneal stromal stem cells exert wound healing effects through the miRNAs that are carried by exosomes [309]. Tao et al. induced an alkali injury in mouse corneas and found that exosomes that were derived from human placental MSCs increased the proliferation and migration of the corneal epithelial cells, and suppressed the inflammation, apoptosis, and angiogenesis at the wound site [314].
In a rabbit model, exosomes from human adipose-derived MSCs (ADSCs) significantly promoted the proliferation and suppressed apoptosis of corneal stromal cells [315]. In addition, the MMP expression was decreased, while extracellular matrix (ECM)-related proteins, such as fibronectin, were increased upon exosomal treatment [315]. These findings support the role of ADSC-derived exosomes in corneal stromal repair and ECM remodeling [315]. Similarly, in a mouse model of dry eye disease, a treatment with ADSC-derived exosomes ameliorated corneal epithelial damage, increased tear production, and decreased inflammatory cytokine proliferation [316]. Thus, ADSC-derived exosomes effectively inhibit NLRP3 inflammasome activation and reduce the corneal surface defects in dry eye disease [316]. In addition, the exosomes that are derived from MSCs and induced pluripotent stem cells (iPSCs) accelerate corneal wound repair [317]. However, iPSCs-derived exosomes exhibited significant effects on the proliferation, migration, and cell cycle promotion of human corneal epithelial cells, thus facilitating tissue regeneration and reducing corneal epithelial defects more effectively than the MSC-derived exosomes [317].
Recently, our group performed the first ever study that explored the role of SEs in corneal wound healing, by using primary corneal stromal cells from healthy (HCFs), type I diabetes mellitus (T1DMs), type II DM (T2DMs), and keratoconus (HKCs) subjects [318]. Scratch and cell migration assays were conducted at 0, 6, 12, 24, and 48 h after the SE stimulation (5 and 25 μg/mL) [318]. After the wound closure, the fibronectin was significantly downregulated in the HKCs, T1DMs, and T2DMs, with 25 μg/mL SE [318]. The HCFs, HKCs, and T2DMs showed a significant upregulation of thrombospondin 1, with 25 μg/mL SE [318]. The cleaved vimentin was significantly upregulated in the HKCs, T1DMs, and T2DMs, with 25 μg/mL SE [318]. These results highlight the potential therapeutic role of SEs in corneal wound healing and establish a strong foundation for future studies.

3.5. Autoimmune Uveitis

Autoimmune uveitis is an inflammation of the uvea, consisting of the iris, ciliary body, and choroid [319]. Its inflammatory response is mediated by pathogenic T cells and interleukins that breach the blood–retinal barrier and damage RPE cells [320,321]. RPE cells have been shown to release exosomes that aid in immunosuppression [322]. For instance, the exosomes from RPE cells that were stimulated with inflammatory cytokines (IL-1β, IFN-γ, and TNF-α) caused the apoptosis of monocytes, while the exosomes from RPE cells that were not stimulated with cytokines induced a pro-inflammatory CD14++CD16+ phenotype in human monocytes [322]. In addition, the stimulated RPE-derived exosomes significantly downregulated the T-cell proliferation, suggesting that RPE-derived exosomes may be able to suppress overactive inflammation in the retina by impeding the proliferation and infiltration of monocytes, as well as other inflammatory cells [322].
Similar to RPE-derived exosomes, MSC-derived exosomes also display immunoprotective effects in autoimmune uveitis [82,323,324]. Using a mixed lymphocyte reaction assay, Shigemoto-Kuroda et al. demonstrated that the exosomes that are secreted by bone-marrow-derived MSCs inhibit the inflammatory response of autoimmune uveitis, by suppressing the activation of antigen-presenting cells and Th1 and Th17 cells [323]. Umbilical cord MSC-derived exosomes were also found to alleviate autoimmune uveitis by blocking the chemoattractive effects of CCL2 and CCL21 on inflammatory cytokines [324]. Additionally, an in vitro study showed that umbilical cord MSC-derived exosomes had a slight inhibitory effect on interphotoreceptor retinoid-binding protein (IRBP)-specific Th17 responses, but significantly inhibited the Th17 responses that are mediated by dendritic cells [82].
Using a mouse model of experimental autoimmune uveitis (EAU), Kang et al. found that IL-35-producing regulatory B-cells (i35-Bregs) secrete exosomes that carry IL-35 [325]. After EAU was induced in mice, a treatment with the IL-35 exosomes resulted in the proliferation of IL-10- and IL-35-secreting T regulatory cells and the attenuation of Th17 responses, while marked choroiditis and photoreceptor cell death were seen in the control mouse eyes [325]. Even more recently, Jiang et al. examined the immunomodulatory role of circulating exosomes that were derived from uveitis rats that had been immunized with interphotoreceptor retinoid-binding protein R16 [326]. The treatment of non-immunized uveitis rats with these circulating exosomes blunted the inflammatory responses of the R16-specific T cells, as evidenced by a reduced IFN-γ and increased IL-10 production [326].

3.6. Uveal Melanoma

Uveal melanoma is a tumor arising from melanocytes [327], which are responsible for producing the pigment of the eye [328]. Uveal melanoma may be located at any point along the uveal tract, but more commonly involves the choroid than the iris and ciliary body [329,330]. It is the most common primary intraocular malignancy in adults, and those affected by the disease are not only at risk of losing their eyesight, but also their life [327,330]. Given the severity and prevalence of uveal melanoma, there have been several studies that have found exosomal biomarker candidates for the disease [257,331,332,333]. Ragusa et al. demonstrated the potential of miR-146a as a biomarker of uveal melanoma by detecting the increased expression of miR-146a in the serum-derived exosomes of uveal melanoma patients [257]. Eldh et al. found that exosomes that were isolated from the liver circulation of uveal melanoma patients contained Melan-A, indicating that these exosomes might be able to travel to the liver vasculature in metastatic uveal melanoma [333]. Interestingly, uveal-melanoma-derived exosomes exhibit a high expression of the proteins that are involved in focal adhesion, endocytosis, and the PI3K-Akt signaling pathway, such as heat shock protein (HSP) 90, HSP70, and integrin V [331], suggesting their involvement in cancer progression. Wroblewska et al. analyzed the proteomic profile of the serum-derived exosomes from primary and metastatic uveal melanoma patients [332] and found that the proteins that were involved in tumor development/metastasis, such as IFN-γ, interleukins 2, 11, and 12, and Pentraxin-3, were significantly elevated in the metastatic uveal melanoma exosomes [332].

3.7. Retinoblastoma

Retinoblastoma is the most common primary intraocular neoplasm in pediatrics, with most tumors occurring in infants and children younger than five years old [334]. The tumor formation occurs when both alleles of the retinoblastoma gene are mutated [334]. Retinoblastoma affects only one eye in approximately 75% of cases, involving spontaneous mutations of the retinoblastoma gene on chromosome 13q14 [334]. Bilateral disease is heritable in an autosomal dominant pattern, due to a germline mutation of the retinoblastoma gene [334]. The study of exosomes to determine the disease mechanisms of retinoblastoma is in its early stages. Castro-Magdonel et al. first examined the miRNA composition of the extracellular vesicles from children that were affected by retinoblastoma, and observed that miRNA-5787 and miRNA-6732-5p were significantly elevated in the extracellular vesicles and plasma of Rb patients [335]. Later, Ravishankar et al. compared the miRNA constituents of exosomes from the retinoblastoma cell lines, WERI-Rb-1 and NCC-Rb-51, versus the exosomes from a control cell line (MIO-M1). Their findings showed that miR-301b-3p and miR-216b-5p were elevated in the exosomes of both Rb cell lines [336]. The exosomes that were derived from the WERI-Rb1 retinoblastoma cells promoted tumor growth by invading the tumor microenvironment, increasing the proportion of tumor-associated macrophages, and decreasing the proportion of natural killer cells [337].

3.8. Proliferative Vitreoretinopathy

Proliferative vitreoretinopathy (PVR) is characterized by the proliferation of vitreous or retinal surface cells that form fibrotic membranes, and can result in tractional retinal detachment [338,339]. Similar to retinoblastoma, exosome-focused studies have only recently gained traction. Since the differentiation of RPE cells into mesenchymal cells via EMT has been associated with the early stages of PVR pathogenesis [338,340], Zhang et al. examined the role of exosomal miRNAs in EMT induction and PVR [341]. An in vitro model of PVR was created by using TGF-β2 to trigger the EMT of RPE cells, and these EMT-induced cells were co-cultured with normal recipient RPE cells [341]. The exosomes from the normal and EMT-induced RPE cells were extracted and analyzed, and 34 differentially expressed miRNAs were detected in the exosomes from the EMT-induced RPE cells [341]. Notably, miR-543 was found in the EMT-induced RPE exosomes and significantly induced the EMT of the recipient RPE cells, highlighting the role of exosomes in triggering PVR via EMT induction [341]. Interestingly, exosomal miR-4488 and miR-1273g-5p have been reported to inhibit the TGF-β2-stimulated EMT in ARPE-19 cells, by downregulating the ATP-binding cassette A4 (ABCA4) [258], which has been reported to be elevated in PVR tissue [342]. Overexpressed ABCA4 can counteract the inhibitory effect of miR-4488 and miR-1273g-5p on the proliferation, migration, and invasion of TGF-β2-stimulated ARPE-19 cells, suggesting that ABCA4-depleting therapies can slow PVR progression [258]. Overall, these studies demonstrate that exosomal miRNAs can both propagate and inhibit PVR fibrotic lesions [258,341].

3.9. Glaucoma

Glaucoma is the leading cause of global irreversible blindness, and the number of people with glaucoma worldwide is expected to increase from 79.6 million in 2020 to 111.8 million by 2040 [343,344]. Glaucoma is characterized by loss of retinal ganglion cells (RGCs), optic nerve atrophy, and the cupping of the optic disc, resulting in visual field defects and eventual blindness [345,346,347]. The primary risk factor for developing glaucoma is increased intraocular pressure (IOP), which is regulated by the outflow of aqueous humor, a fluid that is produced by the ciliary body epithelium and then drained into the systemic circulation via the trabecular meshwork [348,349,350]. The ciliary epithelium secretes exosomes containing RNA to the trabecular meshwork, and the trabecular meshwork releases exosomes back to the ciliary epithelium [351,352]. This exchange of translational signals helps to regulate IOP [351,352]. On the other hand, the disruption of this exosomal transfer may contribute to the development of glaucoma, when the production of aqueous humor is increased and the outflow of aqueous humor is decreased [353,354,355].
The presence of exosomes in aqueous humor has been reported and hypothesized to play a role in the IOP dysregulation in glaucoma [354]. Myocilin, a protein in aqueous humor that is bound to exosomes, may also contribute to the disease progression of glaucoma, since myocilin helps to clear the cell debris within the trabecular meshwork [353]. Mutations in myocilin are seen in glaucoma and lead to a blockage of the aqueous outflow via the trabecular meshwork, increasing the IOP [353]. In a study on the miRNAs that are associated with primary open-angle glaucoma, the levels of miR-182 expression were elevated in the trabecular-meshwork-derived exosomes and aqueous humor, suggesting that miR-182 may be a useful marker of primary open-angle glaucoma [356].
Several studies have revealed the immunoprotective roles of MSC-derived exosomes in glaucoma [357,358]. In a rat model, the addition of bone marrow MSC-derived exosomes to glaucomatous eyes attenuated the RGC atrophy and retinal nerve fiber layer thinning, demonstrating the neuroprotective effects of these exosomes [357]. Mead et al. also reported the neuroprotective effects of bone marrow MSC-derived exosomes in chronic ocular hypertension, showing that exosomes preserved the RGC function for up to 6 months [357]. In addition, exosomes from umbilical cord MSCs promoted the neuroprotection of RGCs in a rat optic nerve crush model [358].
The canonical Wnt signaling pathway is known to be involved in IOP regulation and possibly glaucoma [359,360,361,362]. Multiple studies have reported that non-pigmented ciliary epithelium (NPCE)-derived exosomes altered the canonical Wnt signaling in TM cells, decreasing their beta-catenin protein expression [352,363]. These findings suggest a therapeutic role of NPCE exosomes in glaucoma [352,363]. Specifically, it was shown that the miR-29b that was found in NPCE exosomes was responsible for the downregulation of the Wnt signaling, significantly reducing the levels of collagen III protein expression [351]. It is known that the trabecular meshwork of glaucoma patients is associated with the accumulation of collagen in the ECM and the upregulation of pro-fibrotic factors, such as TGF-β and α-SMA [364,365]. Therefore, upregulating the miR-29b that is carried by exosomes may serve as a useful therapeutic target to suppressing the collagen buildup in the ECM of the trabecular meshwork, normalizing the IOP levels in glaucoma patients [351].
Table 3. Exosomes in ocular diseases.
Table 3. Exosomes in ocular diseases.
DiseaseExosome TypeBiomarkersEffectReference
Diabetic RetinopathyRetinal cellsFibroblast growth factor, TNF-α, angiostatin
miR-20a-3p, miR-20a-5p, miR-20b, VEGF
Pro-inflammatory,
pro-angiogenic
[278,279]
Circular RNA-cPWWP2ARegulates endothelial cell activity via inhibition of miR-579[289]
miR-124-3pAnti-inflammatory[284]
RPEmiR-202-5pAnti-angiogenic [286]
PlasmaIgGDamage to retinal endothelial cells by activating complement[284]
[280]
miR-15aInsulin production by pancreatic beta-cells, oxidative stress in T2D[282]
MSCsmiR-222Retinal repair[287]
Age-Related Macular DegenerationSerummiR-19a, miR-126, miR-410Pro-angiogenic, retinal cell apoptosis [308]
Retinal astroglial cellsEndostatin, MMP-3Anti-angiogenic, inhibit migration of macrophages and endothelial cells[313]
RPEC3, CD63, CD81, LAMP2Drusen production[296]
VEGF-2Pro-angiogenic, retinal endothelial damage[306]
Cathepsin D, cytokeratins 8 and 14Reduce oxidative stress [305]
Retinitis pigmentosaRetinal cellsPARPPhotoreceptor degeneration[294]
RetinoblastomaRetinoblastoma cellsmiR-5787, miR-6732-5p, miR-301b-3p, miR-216b-5p, miR-92a-3pPromote tumor growth and angiogenesis [341,342]
Corneal diseaseCorneal stroma,
Corneal epithelial cells
Fibronectin, TSP-1, α-SMACell migration, myofibroblast differentiation, wound closure[316,319]
Corneal fibroblastsMMP14Pro-angiogenic,
load MMP2 into exosomes
[317]
Limbal stromal cellsKeratin 15Limbal epithelial cell proliferation via Akt phosphorylation[318]
MSCsCol3a1, Acta2, Fibronectin Corneal stromal repair [315,321]
Autoimmune UveitisRPECD14, CD16Anti-inflammatory,
proliferation of IL-10 and T regulatory cells
[328]
Regulatory B-cellsIL-35[331]
SerumRetinoid-binding protein R16[332]
Uveal MelanomaLiver vasculatureMelan-APromote tumor growth and metastasis [339]
Uveal melanoma cellsHSP90, HSP70, integrin V[337]
SerumInterferon-gamma, IL-2, IL-11, IL-12, Pentraxin-3[338]
Proliferative VitreoretinoppathyRPEmiR-543Induce the epithelial–mesenchymal transition (EMT) of recipient RPE cells[347]
miR-4488, miR-1273g-5pInhibit TGF-β2-stimulated EMT in RPE cells by downregulating ABCA4[264]
GlaucomaAqueous humor, Trabecular meshwork,
Non-pigmented ciliary epithelium
Myocilin, miR-182, miR-29bBlockage of aqueous outflow via trabecular meshwork[357,358,359,362]
Recently, Aires et al. found that stressing microglial cells with elevated hydrostatic pressure increased the microglial cell reactivity and retinal cell death, and upregulated exosome release by microglia [366]. Since microglial-cell-mediated inflammation occurs early in glaucoma progression, prior to RGC death, this indicates that the modulation of microglial function can alleviate the RGC degeneration [366].

4. Exosomes as Drug Therapies

The role of exosomes as drug carriers has gained interest due to exosomes’ versatility of function and composition, including bioactive molecules such as RNAs and proteins [11,12,13,14]. In particular, exosomes harbor tetraspanins such as CD63 on their membrane, to which drug molecules can bind with a high affinity, increasing the drug’s efficacy [11,15,367]. In addition, while synthetic drug vehicles suffer from the drawbacks of immunotoxicity and clearance by phagocytes, exosomes can cross the blood–brain barrier and other cell membranes [368,369]. The advantages of using exosomes as drug carriers include low rejection rates, the efficacy of the drug delivery, a prolonged drug activity, and a lower drug dosage required to have a therapeutic effect [3,370]. Exosomes can be utilized for drug delivery by first harvesting the exosomes from the patient, loading the exosomes with therapeutic agents or disease-targeting antibodies, and finally, reinjecting the loaded exosomes back into the patient [371], as displayed in Figure 10.
Exosomes are promising vectors for anti-tumor therapy due to their biocompatibility, low immunogenicity, and innate ability to interact with target cells [372,373,374]. Using exosomes as vehicles for paclitaxel, a drug that inhibits the microtubule assembly of cancer cells [375], has been shown to stabilize the drug concentrations in the plasma, allowing for a prolonged drug bioavailability and an increased drug efficacy [370,375,376]. Engineered exosomes have shown promising results as therapies for multiple cancer types [377,378]. For example, aptamer-modified exosomes targeting prostate cancer cells have effectively inhibited SIRT6 via siRNA delivery, downregulating the Notch and mTOR pathways that are involved in cancer signaling [377]. Exosomes carrying engineered HEK293T cells, which were transduced by a lentiviral vector chimeric gene, were shown to directly bind to the HER2/Neu receptors on breast cancer cells [378]. Upon binding to these HER2/Neu receptors, the exosomes transferred siRNA molecules to the breast cancer cells, downregulating the tumor protein D52 that was overexpressed by the breast cancer cells [378]. Recently, exosomes from normal fibroblasts that were transfected with Epstein–Barr-virus-induced 3 cDNA were electroporated with the siRNA of lymphocyte cytoplasmic protein 1 (LCP1) [379]. These engineered exosomes effectively transferred the siRNA of the LCP1 into OSCC cells, inhibiting their proliferation [379]. Sayyed et al. similarly demonstrated the ability of engineered exosomes to slow cancer progression [380]. Exosomes loaded with the miR-155 inhibitor improve the sensitivity of oral cancer cells to cisplatin, thus reversing miR-155-mediated chemoresistance [380].
Recently, MSC-derived exosomes have shown great promise as tumor-targeted therapies [381]. Using the novel gold nanoparticle labeling of exosomes and computed tomography imaging, Cohen et al. tracked exosomes that were derived from MSCs and the A431 squamous cell carcinoma line in A431 mice [381]. The MSC exosomes demonstrated the ability to selectively target the tumor and distribute throughout the tumor tissue and cytoplasm [381,382]. The MSC exosomes also showed a superior bioavailability, remaining in the bloodstream longer than the A431 exosomes [381]. Similarly, exosomes that were secreted from adipose-derived MSCs effectively inhibited tumor cell metastasis and apoptosis by transferring miR-145 to the breast cancer cells [382].
SEs as drug delivery vehicles have not yet been explored; however, the advantages of saliva as a source of exosomes make SEs a favorable vehicle for drug delivery [30,31,32]. SE collection is convenient and non-invasive, improving patient compliance [31,33]. Additionally, saliva does not coagulate, and SEs have been shown to be stable in biological fluids such as blood and gastric fluid [21,383]. SEs can therefore be easily obtained from the patient [31,33], injected with therapeutic agents, and re-introduced into the patient [384,385]. Using SEs for drug delivery thus has a great therapeutic potential in wide-ranging breakthrough applications, including for autoimmune diseases, neurodegenerative disorders, malignant neoplasms, and many others [386,387].

5. Conclusions

In summary, exosomes have a wide variety of effects on autoimmune diseases [52], neurodegenerative diseases [48,109], and malignant neoplasms [229]. Some notable trends are that MSC-derived exosomes have immunoprotective effects across these three disease categories, slowing the pathogenesis of inflammatory bowel disease [102], Alzheimer’s disease [149,150], oral squamous cell carcinoma [204], and breast cancer [222]. Additionally, SEs have been shown to accelerate the disease propagation in neurodegenerative diseases [22,48], whereas the effects of SEs in systemic autoimmune diseases [47,88] and cancer progression [209,243,255] have only recently been reported, and exhibit both proinflammatory and anti-inflammatory roles. However, the exosome biomarkers in these diseases can serve as useful biomarkers to aid in earlier detection of disease [47,88,209,243,255].
The impact of exosomes on the regulation of ocular diseases is profound and varied [302,313,323,332,366]. The biomarkers that are carried by exosomes possess great potential in monitoring the progression of ocular diseases [258,279,302,325,351]. The biomarkers that are transported by MSC-derived exosomes have exhibited mainly anti-inflammatory effects in diabetic retinopathy [279,280,281] and corneal disease [308,309,314], similar to the effects of MSC-derived exosomes in non-ocular diseases. In contrast, the biomarkers of other exosome types, such as serum-derived exosomes, have been revealed to both alleviate and exacerbate ocular disease pathogenesis [302,326,332].
Together, SEs hold great promise as a future target for numerous therapies, including ocular.

Author Contributions

Conceptualization: D.K. and A.L., resources: D.K. and A.L., writing: original draft: A.L., writing: review and editing: A.L., B.H., P.E., S.E.N. and D.K., visualization: A.L. and B.H., supervision: D.K., project administration: D.K. and A.L., funding acquisition: D.K. All authors have read and agreed to the published version of the manuscript.

Funding

The authors would like to acknowledge the following for their financial support: National Eye Institute, National Institutes of Health; (EY030028-04S1; D.K.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Raposo, G.; Stoorvogel, W. Extracellular vesicles: Exosomes, microvesicles, and friends. J. Cell. Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cocozza, F.; Grisard, E.; Martin-Jaular, L.; Mathieu, M.; Thery, C. SnapShot: Extracellular Vesicles. Cell 2020, 182, 262–262.e1. [Google Scholar] [CrossRef] [PubMed]
  3. Batrakova, E.V.; Kim, M.S. Using exosomes, naturally-equipped nanocarriers, for drug delivery. J. Control. Release 2015, 219, 396–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. O’Brien, K.; Breyne, K.; Ughetto, S.; Laurent, L.C.; Breakefield, X.O. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat. Rev. Mol. Cell Biol. 2020, 21, 585–606. [Google Scholar] [CrossRef] [PubMed]
  5. Thery, 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] [Green Version]
  6. Consortium, E.-T.; Van Deun, J.; Mestdagh, P.; Agostinis, P.; Akay, O.; Anand, S.; Anckaert, J.; Martinez, Z.A.; Baetens, T.; Beghein, E.; et al. EV-TRACK: Transparent reporting and centralizing knowledge in extracellular vesicle research. Nat. Methods 2017, 14, 228–232. [Google Scholar] [CrossRef]
  7. Kowal, J.; Arras, G.; Colombo, M.; Jouve, M.; Morath, J.P.; Primdal-Bengtson, B.; Dingli, F.; Loew, D.; Tkach, M.; Thery, C. Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. USA 2016, 113, E968–E977. [Google Scholar] [CrossRef] [Green Version]
  8. Zhang, L.; Yu, D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 455–468. [Google Scholar] [CrossRef]
  9. Crescitelli, R.; Lasser, C.; Szabo, T.G.; Kittel, A.; Eldh, M.; Dianzani, I.; Buzas, E.I.; Lotvall, J. Distinct RNA profiles in subpopulations of extracellular vesicles: Apoptotic bodies, microvesicles and exosomes. J. Extracell. Vesicles 2013, 2, 20677. [Google Scholar] [CrossRef] [Green Version]
  10. Denzer, K.; Kleijmeer, M.J.; Heijnen, H.F.; Stoorvogel, W.; Geuze, H.J. Exosome: From internal vesicle of the multivesicular body to intercellular signaling device. J. Cell Sci. 2000, 113 Pt 19, 3365–3374. [Google Scholar] [CrossRef]
  11. Thery, C.; Zitvogel, L.; Amigorena, S. Exosomes: Composition, biogenesis and function. Nat. Rev. Immunol. 2002, 2, 569–579. [Google Scholar] [CrossRef]
  12. Zhang, H.G.; Grizzle, W.E. Exosomes: A novel pathway of local and distant intercellular communication that facilitates the growth and metastasis of neoplastic lesions. Am. J. Pathol. 2014, 184, 28–41. [Google Scholar] [CrossRef] [Green Version]
  13. Tan, S.S.; Yin, Y.; Lee, T.; Lai, R.C.; Yeo, R.W.; Zhang, B.; Choo, A.; Lim, S.K. Therapeutic MSC exosomes are derived from lipid raft microdomains in the plasma membrane. J. Extracell. Vesicles 2013, 2, 22614. [Google Scholar] [CrossRef]
  14. van den Boorn, J.G.; Dassler, J.; Coch, C.; Schlee, M.; Hartmann, G. Exosomes as nucleic acid nanocarriers. Adv. Drug. Deliv. Rev. 2013, 65, 331–335. [Google Scholar] [CrossRef]
  15. Palanisamy, V.; Sharma, S.; Deshpande, A.; Zhou, H.; Gimzewski, J.; Wong, D.T. Nanostructural and transcriptomic analyses of human saliva derived exosomes. PLoS ONE 2010, 5, e8577. [Google Scholar] [CrossRef] [Green Version]
  16. Ogawa, Y.; Miura, Y.; Harazono, A.; Kanai-Azuma, M.; Akimoto, Y.; Kawakami, H.; Yamaguchi, T.; Toda, T.; Endo, T.; Tsubuki, M.; et al. Proteomic analysis of two types of exosomes in human whole saliva. Biol. Pharm. Bull. 2011, 34, 13–23. [Google Scholar] [CrossRef] [Green Version]
  17. 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] [Green Version]
  18. Huang, M.B.; Xia, M.; Gao, Z.; Zhou, H.; Liu, M.; Huang, S.; Zhen, R.; Wu, J.Y.; Roth, W.W.; Bond, V.C.; et al. Characterization of Exosomes in Plasma of Patients with Breast, Ovarian, Prostate, Hepatic, Gastric, Colon, and Pancreatic Cancers. J. Cancer Ther. 2019, 10, 382–399. [Google Scholar] [CrossRef] [Green Version]
  19. Hong, C.S.; Funk, S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J. Extracell. Vesicles 2016, 5, 29289. [Google Scholar] [CrossRef]
  20. Michael, A.; Bajracharya, S.D.; Yuen, P.S.; Zhou, H.; Star, R.A.; Illei, G.G.; Alevizos, I. Exosomes from human saliva as a source of microRNA biomarkers. Oral Dis. 2010, 16, 34–38. [Google Scholar] [CrossRef] [Green Version]
  21. Nonaka, T.; Wong, D.T.W. Saliva-Exosomics in Cancer: Molecular Characterization of Cancer-Derived Exosomes in Saliva. Enzymes 2017, 42, 125–151. [Google Scholar] [CrossRef] [PubMed]
  22. Rani, K.; Rastogi, S.; Vishwakarma, P.; Bharti, P.S.; Sharma, V.; Renu, K.; Modi, G.P.; Vishnu, V.Y.; Chatterjee, P.; Dey, A.B.; et al. A novel approach to correlate the salivary exosomes and their protein cargo in the progression of cognitive impairment into Alzheimer’s disease. J. Neurosci. Methods 2021, 347, 108980. [Google Scholar] [CrossRef] [PubMed]
  23. Pisitkun, T.; Shen, R.F.; Knepper, M.A. Identification and proteomic profiling of exosomes in human urine. Proc. Natl. Acad. Sci. USA 2004, 101, 13368–13373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Gheinani, A.H.; Vogeli, M.; Baumgartner, U.; Vassella, E.; Draeger, A.; Burkhard, F.C.; Monastyrskaya, K. Improved isolation strategies to increase the yield and purity of human urinary exosomes for biomarker discovery. Sci. Rep. 2018, 8, 3945. [Google Scholar] [CrossRef] [PubMed]
  25. He, L.; Zhu, D.; Wang, J.; Wu, X. A highly efficient method for isolating urinary exosomes. Int. J. Mol. Med. 2019, 43, 83–90. [Google Scholar] [CrossRef] [Green Version]
  26. Zhu, Q.; Cheng, L.; Deng, C.; Huang, L.; Li, J.; Wang, Y.; Li, M.; Yang, Q.; Dong, X.; Su, J.; et al. The genetic source tracking of human urinary exosomes. Proc. Natl. Acad. Sci. USA 2021, 118, e2108876118. [Google Scholar] [CrossRef]
  27. Keller, S.; Rupp, C.; Stoeck, A.; Runz, S.; Fogel, M.; Lugert, S.; Hager, H.D.; Abdel-Bakky, M.S.; Gutwein, P.; Altevogt, P. CD24 is a marker of exosomes secreted into urine and amniotic fluid. Kidney Int. 2007, 72, 1095–1102. [Google Scholar] [CrossRef] [Green Version]
  28. Dixon, C.L.; Sheller-Miller, S.; Saade, G.R.; Fortunato, S.J.; Lai, A.; Palma, C.; Guanzon, D.; Salomon, C.; Menon, R. Amniotic Fluid Exosome Proteomic Profile Exhibits Unique Pathways of Term and Preterm Labor. Endocrinology 2018, 159, 2229–2240. [Google Scholar] [CrossRef] [Green Version]
  29. Sheller-Miller, S.; Menon, R. Isolation and characterization of human amniotic fluid-derived exosomes. Methods Enzymol. 2020, 645, 181–194. [Google Scholar] [CrossRef]
  30. Cheng, J.; Nonaka, T.; Wong, D.T.W. Salivary Exosomes as Nanocarriers for Cancer Biomarker Delivery. Materials 2019, 12, 654. [Google Scholar] [CrossRef] [Green Version]
  31. Pfaffe, T.; Cooper-White, J.; Beyerlein, P.; Kostner, K.; Punyadeera, C. Diagnostic potential of saliva: Current state and future applications. Clin. Chem. 2011, 57, 675–687. [Google Scholar] [CrossRef] [Green Version]
  32. Wren, M.E.; Shirtcliff, E.A.; Drury, S.S. Not all biofluids are created equal: Chewing over salivary diagnostics and the epigenome. Clin. Ther. 2015, 37, 529–539. [Google Scholar] [CrossRef] [Green Version]
  33. Cecchettini, A.; Finamore, F.; Puxeddu, I.; Ferro, F.; Baldini, C. Salivary extracellular vesicles versus whole saliva: New perspectives for the identification of proteomic biomarkers in Sjogren’s syndrome. Clin. Exp. Rheumatol. 2019, 37 (Suppl. 118), 240–248. [Google Scholar]
  34. Han, Y.; Jia, L.; Zheng, Y.; Li, W. Salivary Exosomes: Emerging Roles in Systemic Disease. Int. J. Biol. Sci. 2018, 14, 633–643. [Google Scholar] [CrossRef] [Green Version]
  35. Helmerhorst, E.J.; Oppenheim, F.G. Saliva: A dynamic proteome. J. Dent. Res. 2007, 86, 680–693. [Google Scholar] [CrossRef]
  36. Gandhi, V.; O’Brien, M.H.; Yadav, S. High-Quality and High-Yield RNA Extraction Method From Whole Human Saliva. Biomark. Insights 2020, 15, 1177271920929705. [Google Scholar] [CrossRef]
  37. Kaczor-Urbanowicz, K.E.; Kim, Y.; Li, F.; Galeev, T.; Kitchen, R.R.; Gerstein, M.; Koyano, K.; Jeong, S.H.; Wang, X.; Elashoff, D.; et al. Novel approaches for bioinformatic analysis of salivary RNA sequencing data for development. Bioinformatics 2018, 34, 1–8. [Google Scholar] [CrossRef] [Green Version]
  38. Bijnsdorp, I.V.; Geldof, A.A.; Lavaei, M.; Piersma, S.R.; van Moorselaar, R.J.; Jimenez, C.R. Exosomal ITGA3 interferes with non-cancerous prostate cell functions and is increased in urine exosomes of metastatic prostate cancer patients. J. Extracell. Vesicles 2013, 2, 22097. [Google Scholar] [CrossRef]
  39. Chiabotto, G.; Gai, C.; Deregibus, M.C.; Camussi, G. Salivary Extracellular Vesicle-Associated exRNA as Cancer Biomarker. Cancers 2019, 11, 891. [Google Scholar] [CrossRef] [Green Version]
  40. van der Lubbe, N.; Jansen, P.M.; Salih, M.; Fenton, R.A.; van den Meiracker, A.H.; Danser, A.H.; Zietse, R.; Hoorn, E.J. The phosphorylated sodium chloride cotransporter in urinary exosomes is superior to prostasin as a marker for aldosteronism. Hypertension 2012, 60, 741–748. [Google Scholar] [CrossRef] [Green Version]
  41. Nilsson, J.; Skog, J.; Nordstrand, A.; Baranov, V.; Mincheva-Nilsson, L.; Breakefield, X.O.; Widmark, A. Prostate cancer-derived urine exosomes: A novel approach to biomarkers for prostate cancer. Br. J. Cancer 2009, 100, 1603–1607. [Google Scholar] [CrossRef] [PubMed]
  42. Zhu, L.; Sun, H.T.; Wang, S.; Huang, S.L.; Zheng, Y.; Wang, C.Q.; Hu, B.Y.; Qin, W.; Zou, T.T.; Fu, Y.; et al. Isolation and characterization of exosomes for cancer research. J. Hematol. Oncol. 2020, 13, 152. [Google Scholar] [CrossRef] [PubMed]
  43. de la Torre, P.; Perez-Lorenzo, M.J.; Alcazar-Garrido, A.; Flores, A.I. Cell-Based Nanoparticles Delivery Systems for Targeted Cancer Therapy: Lessons from Anti-Angiogenesis Treatments. Molecules 2020, 25, 715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Iwai, K.; Minamisawa, T.; Suga, K.; Yajima, Y.; Shiba, K. Isolation of human salivary extracellular vesicles by iodixanol density gradient ultracentrifugation and their characterizations. J. Extracell. Vesicles 2016, 5, 30829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Byun, J.S.; Hong, S.H.; Choi, J.K.; Jung, J.K.; Lee, H.J. Diagnostic profiling of salivary exosomal microRNAs in oral lichen planus patients. Oral Dis. 2015, 21, 987–993. [Google Scholar] [CrossRef]
  46. Ma, H.; Wu, Y.; Yang, H.; Liu, J.; Dan, H.; Zeng, X.; Zhou, Y.; Jiang, L.; Chen, Q. MicroRNAs in oral lichen planus and potential miRNA-mRNA pathogenesis with essential cytokines: A review. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2016, 122, 164–173. [Google Scholar] [CrossRef]
  47. Zheng, X.; Chen, F.; Zhang, Q.; Liu, Y.; You, P.; Sun, S.; Lin, J.; Chen, N. Salivary exosomal PSMA7: A promising biomarker of inflammatory bowel disease. Protein Cell 2017, 8, 686–695. [Google Scholar] [CrossRef] [Green Version]
  48. Cao, Z.; Wu, Y.; Liu, G.; Jiang, Y.; Wang, X.; Wang, Z.; Feng, T. alpha-Synuclein in salivary extracellular vesicles as a potential biomarker of Parkinson’s disease. Neurosci. Lett. 2019, 696, 114–120. [Google Scholar] [CrossRef]
  49. Zeng, Q.; Zhu, Z.; Song, L.; He, Z. Transferred by exosomes-derived MiR-19b-3p targets PTEN to regulate esophageal cancer cell apoptosis, migration and invasion. Biosci. Rep. 2020, 40, BSR20201858. [Google Scholar] [CrossRef]
  50. Theodoraki, M.N.; Yerneni, S.S.; Hoffmann, T.K.; Gooding, W.E.; Whiteside, T.L. Clinical Significance of PD-L1(+) Exosomes in Plasma of Head and Neck Cancer Patients. Clin. Cancer Res. 2018, 24, 896–905. [Google Scholar] [CrossRef] [Green Version]
  51. Han, P.; Bartold, P.M.; Salomon, C.; Ivanovski, S. Salivary Small Extracellular Vesicles Associated miRNAs in Periodontal Status-A Pilot Study. Int. J. Mol. Sci. 2020, 21, 2809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Rui, K.; Hong, Y.; Zhu, Q.; Shi, X.; Xiao, F.; Fu, H.; Yin, Q.; Xing, Y.; Wu, X.; Kong, X.; et al. Olfactory ecto-mesenchymal stem cell-derived exosomes ameliorate murine Sjogren’s syndrome by modulating the function of myeloid-derived suppressor cells. Cell. Mol. Immunol. 2021, 18, 440–451. [Google Scholar] [CrossRef] [PubMed]
  53. Lu, J.; Liu, D.; Tan, Y.; Deng, F.; Li, R. M1 Macrophage exosomes MiR-21a-5p aggravates inflammatory bowel disease through decreasing E-cadherin and subsequent ILC2 activation. J. Cell. Mol. Med. 2021, 25, 3041–3050. [Google Scholar] [CrossRef] [PubMed]
  54. Thorne, J.E.; Jabs, D.A.; Nikolskaia, O.V.; Mimouni, D.; Anhalt, G.J.; Nousari, H.C. Lichen planus and cicatrizing conjunctivitis: Characterization of five cases. Am. J. Ophthalmol. 2003, 136, 239–243. [Google Scholar] [CrossRef]
  55. Li, C.; He, H.; Wang, J.; Xia, X.; Zhang, M.; Wu, Q. Possible roles of exosomal miRNAs in the pathogenesis of oral lichen planus. Am. J. Transl. Res. 2019, 11, 5313–5323. [Google Scholar]
  56. Nogueira, P.A.; Carneiro, S.; Ramos-e-Silva, M. Oral lichen planus: An update on its pathogenesis. Int. J. Dermatol. 2015, 54, 1005–1010. [Google Scholar] [CrossRef]
  57. Huang, Y.Y.; Wang, C.M.; Potenziani, S.; Hsu, S. Lichen planus of the eyelids: A case report and review of the literature. Dermatol. Online J. 2017, 23, 13030. [Google Scholar] [CrossRef]
  58. Rhee, M.K.; Mootha, V.V. Bilateral keratoconjunctivitis associated with lichen planus. Cornea 2004, 23, 100–105. [Google Scholar] [CrossRef]
  59. Gomez-Elizondo, D.E.; Lopez-Martinez, M.; Ruiz-Lozano, R.E.; Valdez-Garcia, J.E.; Hernandez-Camarena, J.C. Corneal perforation associated with isolated ocular lichen planus: A case report. Eur. J. Ophthalmol. 2021, 31, NP9–NP12. [Google Scholar] [CrossRef]
  60. Rozas Munoz, E.; Martinez-Escala, M.E.; Juanpere, N.; Armentia, J.; Pujol, R.M.; Herrero-Gonzalez, J.E. Isolated conjunctival lichen planus: A diagnostic challenge. Arch. Dermatol. 2011, 147, 465–467. [Google Scholar] [CrossRef] [Green Version]
  61. Brewer, J.D.; Ekdawi, N.S.; Torgerson, R.R.; Camilleri, M.J.; Bruce, A.J.; Rogers, R.S., 3rd; Maguire, L.J. Lichen planus and cicatricial conjunctivitis: Disease course and response to therapy of 11 patients. J. Eur. Acad. Dermatol. Venereol. 2011, 25, 100–104. [Google Scholar] [CrossRef]
  62. Peng, Q.; Zhang, J.; Zhou, G. Differentially circulating exosomal microRNAs expression profiling in oral lichen planus. Am. J. Transl. Res. 2018, 10, 2848–2858. [Google Scholar]
  63. Yang, J.Y.; Tan, Y.Q.; Zhou, G. T cell-derived exosomes containing cytokines induced keratinocytes apoptosis in oral lichen planus. Oral Dis. 2021, 28, 682–690. [Google Scholar] [CrossRef]
  64. Yang, J.Y.; Zhang, J.; Lu, R.; Tan, Y.Q.; Du, G.F.; Zhou, G. T cell-derived exosomes induced macrophage inflammatory protein-1alpha/beta drive the trafficking of CD8+ T cells in oral lichen planus. J. Cell. Mol. Med. 2020, 24, 14086–14098. [Google Scholar] [CrossRef]
  65. Peng, Q.; Zhang, J.; Zhou, G. Circulating exosomes regulate T-cell-mediated inflammatory response in oral lichen planus. J. Oral Pathol. Med. 2019, 48, 143–150. [Google Scholar] [CrossRef]
  66. Kononen, E.; Gursoy, M.; Gursoy, U.K. Periodontitis: A Multifaceted Disease of Tooth-Supporting Tissues. J. Clin. Med. 2019, 8, 1135. [Google Scholar] [CrossRef] [Green Version]
  67. Sun, K.T.; Shen, T.C.; Chen, S.C.; Chang, C.L.; Li, C.H.; Li, X.; Palanisamy, K.; Hsia, N.Y.; Chang, W.S.; Tsai, C.W.; et al. Periodontitis and the subsequent risk of glaucoma: Results from the real-world practice. Sci. Rep. 2020, 10, 17568. [Google Scholar] [CrossRef]
  68. Yeh, L.J.; Shen, T.C.; Sun, K.T.; Lin, C.L.; Hsia, N.Y. Periodontitis and Subsequent Risk of Cataract: Results From Real-World Practice. Front. Med. 2022, 9, 721119. [Google Scholar] [CrossRef]
  69. Tang, Y.L.; Shentu, X.C.; Zhao, S.J.; Tang, X.J.; He, L.; Ping, F.Y. Ocular findings in syndromic gingival fibromatosis: A case study and electronic microscopic investigation of lens. Int. J. Ophthalmol. 2014, 7, 574–576. [Google Scholar] [CrossRef]
  70. Guncu, G.N.; Caglayan, F. Resolution of anterior scleritis after periodontal therapy. Eur. J. Dent. 2011, 5, 337–339. [Google Scholar] [CrossRef] [Green Version]
  71. Karesvuo, P.; Gursoy, U.K.; Pussinen, P.J.; Suominen, A.L.; Huumonen, S.; Vesti, E.; Kononen, E. Alveolar bone loss associated with age-related macular degeneration in males. J. Periodontol. 2013, 84, 58–67. [Google Scholar] [CrossRef] [PubMed]
  72. Wagley, S.; Marra, K.V.; Salhi, R.A.; Gautam, S.; Campo, R.; Veale, P.; Veale, J.; Arroyo, J.G. Periodontal disease and age-related macular degeneration: Results from the National Health and Nutrition Examination Survey III. Retina 2015, 35, 982–988. [Google Scholar] [CrossRef] [PubMed]
  73. Zhang, Z.; Shuai, Y.; Zhou, F.; Yin, J.; Hu, J.; Guo, S.; Wang, Y.; Liu, W. PDLSCs Regulate Angiogenesis of Periodontal Ligaments via VEGF Transferred by Exosomes in Periodontitis. Int. J. Med. Sci. 2020, 17, 558–567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Shen, Z.; Kuang, S.; Zhang, Y.; Yang, M.; Qin, W.; Shi, X.; Lin, Z. Chitosan hydrogel incorporated with dental pulp stem cell-derived exosomes alleviates periodontitis in mice via a macrophage-dependent mechanism. Bioact. Mater. 2020, 5, 1113–1126. [Google Scholar] [CrossRef]
  75. Yu, J.; Lin, Y.; Xiong, X.; Li, K.; Yao, Z.; Dong, H.; Jiang, Z.; Yu, D.; Yeung, S.J.; Zhang, H. Detection of Exosomal PD-L1 RNA in Saliva of Patients with Periodontitis. Front. Genet. 2019, 10, 202. [Google Scholar] [CrossRef] [Green Version]
  76. Han, P.; Bartold, P.M.; Salomon, C.; Ivanovski, S. Salivary Outer Membrane Vesicles and DNA Methylation of Small Extracellular Vesicles as Biomarkers for Periodontal Status: A Pilot Study. Int. J. Mol. Sci. 2021, 22, 2423. [Google Scholar] [CrossRef]
  77. Baldini, C.; Ferro, F.; Luciano, N.; Bombardieri, S.; Grossi, E. Artificial neural networks help to identify disease subsets and to predict lymphoma in primary Sjogren’s syndrome. Clin. Exp. Rheumatol. 2018, 36 (Suppl. 112), 137–144. [Google Scholar]
  78. Goules, A.V.; Tzioufas, A.G. Lymphomagenesis in Sjogren’s syndrome: Predictive biomarkers towards precision medicine. Autoimmun. Rev. 2019, 18, 137–143. [Google Scholar] [CrossRef]
  79. Akpek, E.K.; Mathews, P.; Hahn, S.; Hessen, M.; Kim, J.; Grader-Beck, T.; Birnbaum, J.; Baer, A.N. Ocular and systemic morbidity in a longitudinal cohort of Sjogren’s syndrome. Ophthalmology 2015, 122, 56–61. [Google Scholar] [CrossRef]
  80. Li, B.; Xing, Y.; Gan, Y.; He, J.; Hua, H. Labial gland-derived mesenchymal stem cells and their exosomes ameliorate murine Sjogren’s syndrome by modulating the balance of Treg and Th17 cells. Stem Cell Res. Ther. 2021, 12, 478. [Google Scholar] [CrossRef]
  81. Cortes-Troncoso, J.; Jang, S.I.; Perez, P.; Hidalgo, J.; Ikeuchi, T.; Greenwell-Wild, T.; Warner, B.M.; Moutsopoulos, N.M.; Alevizos, I. T cell exosome-derived miR-142-3p impairs glandular cell function in Sjogren’s syndrome. JCI Insight 2020, 5, e133497. [Google Scholar] [CrossRef]
  82. Li, N.; Zhao, L.; Wei, Y.; Ea, V.L.; Nian, H.; Wei, R. Recent advances of exosomes in immune-mediated eye diseases. Stem Cell Res. Ther. 2019, 10, 278. [Google Scholar] [CrossRef] [Green Version]
  83. Liu, Y.; Li, C.; Wang, S.; Guo, J.; Guo, J.; Fu, J.; Ren, L.; An, Y.; He, J.; Li, Z. Human umbilical cord mesenchymal stem cells confer potent immunosuppressive effects in Sjogren’s syndrome by inducing regulatory T cells. Mod. Rheumatol. 2021, 31, 186–196. [Google Scholar] [CrossRef]
  84. Gallo, A.; Jang, S.I.; Ong, H.L.; Perez, P.; Tandon, M.; Ambudkar, I.; Illei, G.; Alevizos, I. Targeting the Ca2+ Sensor STIM1 by Exosomal Transfer of Ebv-miR-BART13-3p is Associated with Sjogren’s Syndrome. EBioMedicine 2016, 10, 216–226. [Google Scholar] [CrossRef] [Green Version]
  85. Alevizos, I.; Alexander, S.; Turner, R.J.; Illei, G.G. MicroRNA expression profiles as biomarkers of minor salivary gland inflammation and dysfunction in Sjogren’s syndrome. Arthritis Rheum. 2011, 63, 535–544. [Google Scholar] [CrossRef] [Green Version]
  86. Hu, S.; Wang, J.; Meijer, J.; Ieong, S.; Xie, Y.; Yu, T.; Zhou, H.; Henry, S.; Vissink, A.; Pijpe, J.; et al. Salivary proteomic and genomic biomarkers for primary Sjogren’s syndrome. Arthritis Rheum. 2007, 56, 3588–3600. [Google Scholar] [CrossRef] [Green Version]
  87. Kapsogeorgou, E.K.; Abu-Helu, R.F.; Moutsopoulos, H.M.; Manoussakis, M.N. Salivary gland epithelial cell exosomes: A source of autoantigenic ribonucleoproteins. Arthritis Rheum. 2005, 52, 1517–1521. [Google Scholar] [CrossRef]
  88. Aqrawi, L.A.; Galtung, H.K.; Vestad, B.; Ovstebo, R.; Thiede, B.; Rusthen, S.; Young, A.; Guerreiro, E.M.; Utheim, T.P.; Chen, X.; et al. Identification of potential saliva and tear biomarkers in primary Sjogren’s syndrome, utilising the extraction of extracellular vesicles and proteomics analysis. Arthritis Res. Ther. 2017, 19, 14. [Google Scholar] [CrossRef] [Green Version]
  89. Feuerstein, J.D.; Cheifetz, A.S. Crohn Disease: Epidemiology, Diagnosis, and Management. Mayo Clin. Proc. 2017, 92, 1088–1103. [Google Scholar] [CrossRef] [Green Version]
  90. Baumgart, D.C.; Carding, S.R. Inflammatory bowel disease: Cause and immunobiology. Lancet 2007, 369, 1627–1640. [Google Scholar] [CrossRef]
  91. Podolsky, D.K. Inflammatory bowel disease (1). N. Engl. J. Med. 1991, 325, 928–937. [Google Scholar] [CrossRef] [PubMed]
  92. Soukiasian, S.H.; Foster, C.S.; Raizman, M.B. Treatment strategies for scleritis and uveitis associated with inflammatory bowel disease. Am. J. Ophthalmol. 1994, 118, 601–611. [Google Scholar] [CrossRef] [PubMed]
  93. Calvo, P.; Pablo, L. Managing IBD outside the gut: Ocular manifestations. Dig. Dis. 2013, 31, 229–232. [Google Scholar] [CrossRef] [PubMed]
  94. Mintz, R.; Feller, E.R.; Bahr, R.L.; Shah, S.A. Ocular manifestations of inflammatory bowel disease. Inflamm. Bowel Dis. 2004, 10, 135–139. [Google Scholar] [CrossRef] [PubMed]
  95. Troncoso, L.L.; Biancardi, A.L.; de Moraes, H.V., Jr.; Zaltman, C. Ophthalmic manifestations in patients with inflammatory bowel disease: A review. World J. Gastroenterol. 2017, 23, 5836–5848. [Google Scholar] [CrossRef]
  96. Bernstein, C.N.; Blanchard, J.F.; Rawsthorne, P.; Yu, N. The prevalence of extraintestinal diseases in inflammatory bowel disease: A population-based study. Am. J. Gastroenterol. 2001, 96, 1116–1122. [Google Scholar] [CrossRef]
  97. Mady, R.; Grover, W.; Butrus, S. Ocular complications of inflammatory bowel disease. Sci. World J. 2015, 2015, 438402. [Google Scholar] [CrossRef] [Green Version]
  98. Orchard, T.R.; Chua, C.N.; Ahmad, T.; Cheng, H.; Welsh, K.I.; Jewell, D.P. Uveitis and erythema nodosum in inflammatory bowel disease: Clinical features and the role of HLA genes. Gastroenterology 2002, 123, 714–718. [Google Scholar] [CrossRef]
  99. Lakatos, L.; Pandur, T.; David, G.; Balogh, Z.; Kuronya, P.; Tollas, A.; Lakatos, P.L. Association of extraintestinal manifestations of inflammatory bowel disease in a province of western Hungary with disease phenotype: Results of a 25-year follow-up study. World J. Gastroenterol. 2003, 9, 2300–2307. [Google Scholar] [CrossRef]
  100. Wu, Y.; Qiu, W.; Xu, X.; Kang, J.; Wang, J.; Wen, Y.; Tang, X.; Yan, Y.; Qian, H.; Zhang, X.; et al. Exosomes derived from human umbilical cord mesenchymal stem cells alleviate inflammatory bowel disease in mice through ubiquitination. Am. J. Transl. Res. 2018, 10, 2026–2036. [Google Scholar]
  101. Yang, S.; Liang, X.; Song, J.; Li, C.; Liu, A.; Luo, Y.; Ma, H.; Tan, Y.; Zhang, X. A novel therapeutic approach for inflammatory bowel disease by exosomes derived from human umbilical cord mesenchymal stem cells to repair intestinal barrier via TSG-6. Stem Cell Res. Ther. 2021, 12, 315. [Google Scholar] [CrossRef]
  102. Yu, H.; Yang, X.; Xiao, X.; Xu, M.; Yang, Y.; Xue, C.; Li, X.; Wang, S.; Zhao, R.C. Human Adipose Mesenchymal Stem Cell-derived Exosomes Protect Mice from DSS-Induced Inflammatory Bowel Disease by Promoting Intestinal-stem-cell and Epithelial Regeneration. Aging Dis. 2021, 12, 1423–1437. [Google Scholar] [CrossRef]
  103. Mao, F.; Wu, Y.; Tang, X.; Kang, J.; Zhang, B.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Relieve Inflammatory Bowel Disease in Mice. Biomed. Res. Int. 2017, 2017, 5356760. [Google Scholar] [CrossRef] [Green Version]
  104. Cleynen, I.; Vazeille, E.; Artieda, M.; Verspaget, H.W.; Szczypiorska, M.; Bringer, M.A.; Lakatos, P.L.; Seibold, F.; Parnell, K.; Weersma, R.K.; et al. Genetic and microbial factors modulating the ubiquitin proteasome system in inflammatory bowel disease. Gut 2014, 63, 1265–1274. [Google Scholar] [CrossRef]
  105. Hetzenecker, A.M.; Seidl, M.C.; Kosovac, K.; Herfarth, H.; Kellermeier, S.; Obermeier, F.; Falk, W.; Schoelmerich, J.; Hausmann, M.; Rogler, G. Downregulation of the ubiquitin-proteasome system in normal colonic macrophages and reinduction in inflammatory bowel disease. Digestion 2012, 86, 34–47. [Google Scholar] [CrossRef] [Green Version]
  106. Wang, G.; Yuan, J.; Cai, X.; Xu, Z.; Wang, J.; Ocansey, D.K.W.; Yan, Y.; Qian, H.; Zhang, X.; Xu, W.; et al. HucMSC-exosomes carrying miR-326 inhibit neddylation to relieve inflammatory bowel disease in mice. Clin. Transl. Med. 2020, 10, e113. [Google Scholar] [CrossRef]
  107. Wong, W.Y.; Lee, M.M.; Chan, B.D.; Kam, R.K.; Zhang, G.; Lu, A.P.; Tai, W.C. Proteomic profiling of dextran sulfate sodium induced acute ulcerative colitis mice serum exosomes and their immunomodulatory impact on macrophages. Proteomics 2016, 16, 1131–1145. [Google Scholar] [CrossRef]
  108. Liao, F.; Lu, X.; Dong, W. Exosomes derived from T regulatory cells relieve inflammatory bowel disease by transferring miR-195a-3p. IUBMB Life 2020, 72, 2591–2600. [Google Scholar] [CrossRef]
  109. Apodaca, L.A.; Baddour, A.A.D.; Garcia, C., Jr.; Alikhani, L.; Giedzinski, E.; Ru, N.; Agrawal, A.; Acharya, M.M.; Baulch, J.E. Human neural stem cell-derived extracellular vesicles mitigate hallmarks of Alzheimer’s disease. Alzheimers Res. Ther. 2021, 13, 57. [Google Scholar] [CrossRef]
  110. Chen, H.X.; Liang, F.C.; Gu, P.; Xu, B.L.; Xu, H.J.; Wang, W.T.; Hou, J.Y.; Xie, D.X.; Chai, X.Q.; An, S.J. Exosomes derived from mesenchymal stem cells repair a Parkinson’s disease model by inducing autophagy. Cell Death Dis. 2020, 11, 288. [Google Scholar] [CrossRef]
  111. Zou, J.; Guo, Y.; Wei, L.; Yu, F.; Yu, B.; Xu, A. Long Noncoding RNA POU3F3 and alpha-Synuclein in Plasma L1CAM Exosomes Combined with beta-Glucocerebrosidase Activity: Potential Predictors of Parkinson’s Disease. Neurotherapeutics 2020, 17, 1104–1119. [Google Scholar] [CrossRef] [PubMed]
  112. Perl, D.P. Neuropathology of Alzheimer’s disease. Mt. Sinai J. Med. 2010, 77, 32–42. [Google Scholar] [CrossRef] [PubMed]
  113. Jack, C.R., Jr.; Bennett, D.A.; Blennow, K.; Carrillo, M.C.; Dunn, B.; Haeberlein, S.B.; Holtzman, D.M.; Jagust, W.; Jessen, F.; Karlawish, J.; et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement. 2018, 14, 535–562. [Google Scholar] [CrossRef]
  114. Kumar, A.; Singh, A.; Ekavali. A review on Alzheimer’s disease pathophysiology and its management: An update. Pharmacol. Rep. 2015, 67, 195–203. [Google Scholar] [CrossRef] [PubMed]
  115. Tiwari, S.; Atluri, V.; Kaushik, A.; Yndart, A.; Nair, M. Alzheimer’s disease: Pathogenesis, diagnostics, and therapeutics. Int. J. Nanomed. 2019, 14, 5541–5554. [Google Scholar] [CrossRef] [Green Version]
  116. Samanta, S.; Rajasingh, S.; Drosos, N.; Zhou, Z.; Dawn, B.; Rajasingh, J. Exosomes: New molecular targets of diseases. Acta Pharmacol. Sin. 2018, 39, 501–513. [Google Scholar] [CrossRef]
  117. D’Anca, M.; Fenoglio, C.; Serpente, M.; Arosio, B.; Cesari, M.; Scarpini, E.A.; Galimberti, D. Exosome Determinants of Physiological Aging and Age-Related Neurodegenerative Diseases. Front. Aging Neurosci. 2019, 11, 232. [Google Scholar] [CrossRef] [Green Version]
  118. Wang, J.; Gu, B.J.; Masters, C.L.; Wang, Y.J. A systemic view of Alzheimer disease—Insights from amyloid-beta metabolism beyond the brain. Nat. Rev. Neurol. 2017, 13, 703. [Google Scholar] [CrossRef] [Green Version]
  119. Zhao, Y.; Bhattacharjee, S.; Jones, B.M.; Hill, J.M.; Clement, C.; Sambamurti, K.; Dua, P.; Lukiw, W.J. Beta-Amyloid Precursor Protein (betaAPP) Processing in Alzheimer’s Disease (AD) and Age-Related Macular Degeneration (AMD). Mol. Neurobiol. 2015, 52, 533–544. [Google Scholar] [CrossRef] [Green Version]
  120. Ding, J.D.; Lin, J.; Mace, B.E.; Herrmann, R.; Sullivan, P.; Bowes Rickman, C. Targeting age-related macular degeneration with Alzheimer’s disease based immunotherapies: Anti-amyloid-beta antibody attenuates pathologies in an age-related macular degeneration mouse model. Vis. Res. 2008, 48, 339–345. [Google Scholar] [CrossRef] [Green Version]
  121. Ukalovic, K.; Cao, S.; Lee, S.; Tang, Q.; Beg, M.F.; Sarunic, M.V.; Hsiung, G.R.; Mackenzie, I.R.; Hirsch-Reinshagen, V.; Cui, J.Z.; et al. Drusen in the Peripheral Retina of the Alzheimer’s Eye. Curr. Alzheimer Res. 2018, 15, 743–750. [Google Scholar] [CrossRef]
  122. Tsai, D.C.; Chen, S.J.; Huang, C.C.; Yuan, M.K.; Leu, H.B. Age-Related Macular Degeneration and Risk of Degenerative Dementia among the Elderly in Taiwan: A Population-Based Cohort Study. Ophthalmology 2015, 122, 2327–2335.e2. [Google Scholar] [CrossRef]
  123. Parisi, V. Correlation between morphological and functional retinal impairment in patients affected by ocular hypertension, glaucoma, demyelinating optic neuritis and Alzheimer’s disease. Semin. Ophthalmol. 2003, 18, 50–57. [Google Scholar] [CrossRef]
  124. Cipollini, V.; Abdolrahimzadeh, S.; Troili, F.; De Carolis, A.; Calafiore, S.; Scuderi, L.; Giubilei, F.; Scuderi, G. Neurocognitive Assessment and Retinal Thickness Alterations in Alzheimer Disease: Is There a Correlation? J. Neuroophthalmol. 2020, 40, 370–377. [Google Scholar] [CrossRef]
  125. Iseri, P.K.; Altinas, O.; Tokay, T.; Yuksel, N. Relationship between cognitive impairment and retinal morphological and visual functional abnormalities in Alzheimer disease. J. Neuroophthalmol. 2006, 26, 18–24. [Google Scholar] [CrossRef]
  126. Kirbas, S.; Turkyilmaz, K.; Anlar, O.; Tufekci, A.; Durmus, M. Retinal nerve fiber layer thickness in patients with Alzheimer disease. J. Neuroophthalmol. 2013, 33, 58–61. [Google Scholar] [CrossRef] [Green Version]
  127. Risacher, S.L.; Wudunn, D.; Pepin, S.M.; MaGee, T.R.; McDonald, B.C.; Flashman, L.A.; Wishart, H.A.; Pixley, H.S.; Rabin, L.A.; Pare, N.; et al. Visual contrast sensitivity in Alzheimer’s disease, mild cognitive impairment, and older adults with cognitive complaints. Neurobiol. Aging 2013, 34, 1133–1144. [Google Scholar] [CrossRef] [Green Version]
  128. Risacher, S.L.; WuDunn, D.; Tallman, E.F.; West, J.D.; Gao, S.; Farlow, M.R.; Brosch, J.R.; Apostolova, L.G.; Saykin, A.J. Visual contrast sensitivity is associated with the presence of cerebral amyloid and tau deposition. Brain Commun. 2020, 2, fcaa019. [Google Scholar] [CrossRef]
  129. Gilmore, G.C.; Groth, K.E.; Thomas, C.W. Stimulus contrast and word reading speed in Alzheimer’s disease. Exp. Aging Res. 2005, 31, 15–33. [Google Scholar] [CrossRef]
  130. Fernandez, G.; Mandolesi, P.; Rotstein, N.P.; Colombo, O.; Agamennoni, O.; Politi, L.E. Eye movement alterations during reading in patients with early Alzheimer disease. Investig. Ophthalmol. Vis. Sci. 2013, 54, 8345–8352. [Google Scholar] [CrossRef] [Green Version]
  131. Garbutt, S.; Matlin, A.; Hellmuth, J.; Schenk, A.K.; Johnson, J.K.; Rosen, H.; Dean, D.; Kramer, J.; Neuhaus, J.; Miller, B.L.; et al. Oculomotor function in frontotemporal lobar degeneration, related disorders and Alzheimer’s disease. Brain 2008, 131, 1268–1281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Shafiq-Antonacci, R.; Maruff, P.; Masters, C.; Currie, J. Spectrum of saccade system function in Alzheimer disease. Arch. Neurol. 2003, 60, 1272–1278. [Google Scholar] [CrossRef] [Green Version]
  133. Byun, M.S.; Park, S.W.; Lee, J.H.; Yi, D.; Jeon, S.Y.; Choi, H.J.; Joung, H.; Ghim, U.H.; Park, U.C.; Kim, Y.K.; et al. Association of Retinal Changes With Alzheimer Disease Neuroimaging Biomarkers in Cognitively Normal Individuals. JAMA Ophthalmol. 2021, 139, 548–556. [Google Scholar] [CrossRef] [PubMed]
  134. Lee, C.S.; Apte, R.S. Retinal Biomarkers of Alzheimer Disease. Am. J. Ophthalmol. 2020, 218, 337–341. [Google Scholar] [CrossRef] [PubMed]
  135. Tang, M.Y.; Blazes, M.S.; Lee, C.S. Imaging Amyloid and Tau in the Retina: Current Research and Future Directions. J. Neuroophthalmol. 2023. [Google Scholar] [CrossRef]
  136. Winston, C.N.; Goetzl, E.J.; Schwartz, J.B.; Elahi, F.M.; Rissman, R.A. Complement protein levels in plasma astrocyte-derived exosomes are abnormal in conversion from mild cognitive impairment to Alzheimer’s disease dementia. Alzheimers Dement. 2019, 11, 61–66. [Google Scholar] [CrossRef]
  137. Goetzl, E.J.; Mustapic, M.; Kapogiannis, D.; Eitan, E.; Lobach, I.V.; Goetzl, L.; Schwartz, J.B.; Miller, B.L. Cargo proteins of plasma astrocyte-derived exosomes in Alzheimer’s disease. FASEB J. 2016, 30, 3853–3859. [Google Scholar] [CrossRef] [Green Version]
  138. Saman, S.; Lee, N.C.; Inoyo, I.; Jin, J.; Li, Z.; Doyle, T.; McKee, A.C.; Hall, G.F. Proteins recruited to exosomes by tau overexpression implicate novel cellular mechanisms linking tau secretion with Alzheimer’s disease. J. Alzheimers Dis. 2014, 40 (Suppl. 1), S47–S70. [Google Scholar] [CrossRef]
  139. Fiandaca, M.S.; Kapogiannis, D.; Mapstone, M.; Boxer, A.; Eitan, E.; Schwartz, J.B.; Abner, E.L.; Petersen, R.C.; Federoff, H.J.; Miller, B.L.; et al. Identification of preclinical Alzheimer’s disease by a profile of pathogenic proteins in neurally derived blood exosomes: A case-control study. Alzheimers Dement. 2015, 11, 600–607.e1. [Google Scholar] [CrossRef] [Green Version]
  140. Rosas-Hernandez, H.; Cuevas, E.; Raymick, J.B.; Robinson, B.L.; Ali, S.F.; Hanig, J.; Sarkar, S. Characterization of Serum Exosomes from a Transgenic Mouse Model of Alzheimer’s Disease. Curr. Alzheimer Res. 2019, 16, 388–395. [Google Scholar] [CrossRef]
  141. Cole, S.L.; Vassar, R. The Alzheimer’s disease beta-secretase enzyme, BACE1. Mol. Neurodegener. 2007, 2, 22. [Google Scholar] [CrossRef] [Green Version]
  142. Yan, R.; Vassar, R. Targeting the beta secretase BACE1 for Alzheimer’s disease therapy. Lancet Neurol. 2014, 13, 319–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Chiarini, A.; Armato, U.; Gardenal, E.; Gui, L.; Dal Pra, I. Amyloid beta-Exposed Human Astrocytes Overproduce Phospho-Tau and Overrelease It within Exosomes, Effects Suppressed by Calcilytic NPS 2143-Further Implications for Alzheimer’s Therapy. Front. Neurosci. 2017, 11, 217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Sun, R.; Wang, H.; Shi, Y.; Gao, D.; Sun, Z.; Chen, Z.; Jiang, H.; Zhang, J. A Pilot Study of Urinary Exosomes in Alzheimer’s Disease. Neurodegener. Dis. 2019, 19, 184–191. [Google Scholar] [CrossRef] [PubMed]
  145. Aulston, B.; Liu, Q.; Mante, M.; Florio, J.; Rissman, R.A.; Yuan, S.H. Extracellular Vesicles Isolated from Familial Alzheimer’s Disease Neuronal Cultures Induce Aberrant Tau Phosphorylation in the Wild-Type Mouse Brain. J. Alzheimers Dis. 2019, 72, 575–585. [Google Scholar] [CrossRef]
  146. Yuyama, K.; Sun, H.; Usuki, S.; Sakai, S.; Hanamatsu, H.; Mioka, T.; Kimura, N.; Okada, M.; Tahara, H.; Furukawa, J.; et al. A potential function for neuronal exosomes: Sequestering intracerebral amyloid-beta peptide. FEBS Lett. 2015, 589, 84–88. [Google Scholar] [CrossRef]
  147. Yuyama, K.; Sun, H.; Mitsutake, S.; Igarashi, Y. Sphingolipid-modulated exosome secretion promotes clearance of amyloid-beta by microglia. J. Biol. Chem. 2012, 287, 10977–10989. [Google Scholar] [CrossRef] [Green Version]
  148. 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.; et al. Exosomes neutralize synaptic-plasticity-disrupting activity of Abeta assemblies in vivo. Mol. Brain 2013, 6, 47. [Google Scholar] [CrossRef] [Green Version]
  149. Zhdanova, D.Y.; Poltavtseva, R.A.; Svirshchevskaya, E.V.; Bobkova, N.V. Effect of Intranasal Administration of Multipotent Mesenchymal Stromal Cell Exosomes on Memory of Mice in Alzheimer’s Disease Model. Bull. Exp. Biol. Med. 2021, 170, 575–582. [Google Scholar] [CrossRef]
  150. Wang, X.; Yang, G. Bone marrow mesenchymal stem cells-derived exosomes reduce Abeta deposition and improve cognitive function recovery in mice with Alzheimer’s disease by activating sphingosine kinase/sphingosine-1-phosphate signaling pathway. Cell. Biol. Int. 2021, 45, 775–784. [Google Scholar] [CrossRef]
  151. Chen, Y.A.; Lu, C.H.; Ke, C.C.; Chiu, S.J.; Jeng, F.S.; Chang, C.W.; Yang, B.H.; Liu, R.S. Mesenchymal Stem Cell-Derived Exosomes Ameliorate Alzheimer’s Disease Pathology and Improve Cognitive Deficits. Biomedicines 2021, 9, 594. [Google Scholar] [CrossRef]
  152. Xie, Z.H.; Liu, Z.; Zhang, X.R.; Yang, H.; Wei, L.F.; Wang, Y.; Xu, S.L.; Sun, L.; Lai, C.; Bi, J.Z.; et al. Wharton’s Jelly-derived mesenchymal stem cells alleviate memory deficits and reduce amyloid-beta deposition in an APP/PS1 transgenic mouse model. Clin. Exp. Med. 2016, 16, 89–98. [Google Scholar] [CrossRef]
  153. Ding, M.; Shen, Y.; Wang, P.; Xie, Z.; Xu, S.; Zhu, Z.; Wang, Y.; Lyu, Y.; Wang, D.; Xu, L.; et al. Exosomes Isolated From Human Umbilical Cord Mesenchymal Stem Cells Alleviate Neuroinflammation and Reduce Amyloid-Beta Deposition by Modulating Microglial Activation in Alzheimer’s Disease. Neurochem. Res. 2018, 43, 2165–2177. [Google Scholar] [CrossRef]
  154. Portelius, E.; Dean, R.A.; Andreasson, U.; Mattsson, N.; Westerlund, A.; Olsson, M.; Demattos, R.B.; Racke, M.M.; Zetterberg, H.; May, P.C.; et al. beta-site amyloid precursor protein-cleaving enzyme 1(BACE1) inhibitor treatment induces Abeta5-X peptides through alternative amyloid precursor protein cleavage. Alzheimers Res. Ther. 2014, 6, 75. [Google Scholar] [CrossRef] [Green Version]
  155. Anderson, K.W.; Chen, J.; Wang, M.; Mast, N.; Pikuleva, I.A.; Turko, I.V. Quantification of histone deacetylase isoforms in human frontal cortex, human retina, and mouse brain. PLoS ONE 2015, 10, e0126592. [Google Scholar] [CrossRef] [Green Version]
  156. Atik, A.; Stewart, T.; Zhang, J. Alpha-Synuclein as a Biomarker for Parkinson’s Disease. Brain Pathol. 2016, 26, 410–418. [Google Scholar] [CrossRef]
  157. Devic, I.; Hwang, H.; Edgar, J.S.; Izutsu, K.; Presland, R.; Pan, C.; Goodlett, D.R.; Wang, Y.; Armaly, J.; Tumas, V.; et al. Salivary alpha-synuclein and DJ-1: Potential biomarkers for Parkinson’s disease. Brain 2011, 134, e178. [Google Scholar] [CrossRef] [Green Version]
  158. Greenland, J.C.; Barker, R.A. The Differential Diagnosis of Parkinson’s Disease. In Parkinson’s Disease: Pathogenesis and Clinical Aspects; Stoker, T.B., Greenland, J.C., Eds.; Exon Publications: Brisbane, Australia, 2018. [Google Scholar] [CrossRef]
  159. Massano, J.; Bhatia, K.P. Clinical approach to Parkinson’s disease: Features, diagnosis, and principles of management. Cold Spring Harb. Perspect. Med. 2012, 2, a008870. [Google Scholar] [CrossRef]
  160. Corin, M.S.; Elizan, T.S.; Bender, M.B. Oculomotor function in patients with Parkinson’s disease. J. Neurol. Sci. 1972, 15, 251–265. [Google Scholar] [CrossRef]
  161. Shibasaki, H.; Tsuji, S.; Kuroiwa, Y. Oculomotor abnormalities in Parkinson’s disease. Arch. Neurol. 1979, 36, 360–364. [Google Scholar] [CrossRef]
  162. Almer, Z.; Klein, K.S.; Marsh, L.; Gerstenhaber, M.; Repka, M.X. Ocular motor and sensory function in Parkinson’s disease. Ophthalmology 2012, 119, 178–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Hanuska, J.; Bonnet, C.; Rusz, J.; Sieger, T.; Jech, R.; Rivaud-Pechoux, S.; Vidailhet, M.; Gaymard, B.; Ruzicka, E. Fast vergence eye movements are disrupted in Parkinson’s disease: A video-oculography study. Park. Relat. Disord. 2015, 21, 797–799. [Google Scholar] [CrossRef] [PubMed]
  164. Nowacka, B.; Lubinski, W.; Honczarenko, K.; Potemkowski, A.; Safranow, K. Ophthalmological features of Parkinson disease. Med. Sci. Monit. 2014, 20, 2243–2249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Archibald, N.K.; Clarke, M.P.; Mosimann, U.P.; Burn, D.J. Visual symptoms in Parkinson’s disease and Parkinson’s disease dementia. Mov. Disord. 2011, 26, 2387–2395. [Google Scholar] [CrossRef] [PubMed]
  166. Davidsdottir, S.; Cronin-Golomb, A.; Lee, A. Visual and spatial symptoms in Parkinson’s disease. Vis. Res. 2005, 45, 1285–1296. [Google Scholar] [CrossRef] [Green Version]
  167. Nebe, A.; Ebersbach, G. Selective diplopia in Parkinson’s disease: A special subtype of visual hallucination? Mov. Disord. 2007, 22, 1175–1178. [Google Scholar] [CrossRef]
  168. Repka, M.X.; Claro, M.C.; Loupe, D.N.; Reich, S.G. Ocular motility in Parkinson’s disease. J. Pediatr. Ophthalmol. Strabismus 1996, 33, 144–147. [Google Scholar] [CrossRef]
  169. Tamer, C.; Melek, I.M.; Duman, T.; Oksuz, H. Tear film tests in Parkinson’s disease patients. Ophthalmology 2005, 112, 1795. [Google Scholar] [CrossRef]
  170. Reddy, V.C.; Patel, S.V.; Hodge, D.O.; Leavitt, J.A. Corneal sensitivity, blink rate, and corneal nerve density in progressive supranuclear palsy and Parkinson disease. Cornea 2013, 32, 631–635. [Google Scholar] [CrossRef]
  171. Borm, C.; Smilowska, K.; de Vries, N.M.; Bloem, B.R.; Theelen, T. How I do it: The Neuro-Ophthalmological Assessment in Parkinson’s Disease. J. Park. Dis. 2019, 9, 427–435. [Google Scholar] [CrossRef] [Green Version]
  172. Bayer, A.U.; Keller, O.N.; Ferrari, F.; Maag, K.P. Association of glaucoma with neurodegenerative diseases with apoptotic cell death: Alzheimer’s disease and Parkinson’s disease. Am. J. Ophthalmol. 2002, 133, 135–137. [Google Scholar] [CrossRef]
  173. Tsironi, E.E.; Dastiridou, A.; Katsanos, A.; Dardiotis, E.; Veliki, S.; Patramani, G.; Zacharaki, F.; Ralli, S.; Hadjigeorgiou, G.M. Perimetric and retinal nerve fiber layer findings in patients with Parkinson’s disease. BMC Ophthalmol. 2012, 12, 54. [Google Scholar] [CrossRef] [Green Version]
  174. Grey, M.; Dunning, C.J.; Gaspar, R.; Grey, C.; Brundin, P.; Sparr, E.; Linse, S. Acceleration of alpha-synuclein aggregation by exosomes. J. Biol. Chem. 2015, 290, 2969–2982. [Google Scholar] [CrossRef] [Green Version]
  175. Shi, M.; Liu, C.; Cook, T.J.; Bullock, K.M.; Zhao, Y.; Ginghina, C.; Li, Y.; Aro, P.; Dator, R.; He, C.; et al. Plasma exosomal alpha-synuclein is likely CNS-derived and increased in Parkinson’s disease. Acta Neuropathol. 2014, 128, 639–650. [Google Scholar] [CrossRef] [PubMed]
  176. Majbour, N.K.; Aasly, J.O.; Hustad, E.; Thomas, M.A.; Vaikath, N.N.; Elkum, N.; van de Berg, W.D.J.; Tokuda, T.; Mollenhauer, B.; Berendse, H.W.; et al. CSF total and oligomeric alpha-Synuclein along with TNF-alpha as risk biomarkers for Parkinson’s disease: A study in LRRK2 mutation carriers. Transl. Neurodegener. 2020, 9, 15. [Google Scholar] [CrossRef] [PubMed]
  177. Stuendl, A.; Kunadt, M.; Kruse, N.; Bartels, C.; Moebius, W.; Danzer, K.M.; Mollenhauer, B.; Schneider, A. Induction of alpha-synuclein aggregate formation by CSF exosomes from patients with Parkinson’s disease and dementia with Lewy bodies. Brain 2016, 139, 481–494. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Guo, M.; Wang, J.; Zhao, Y.; Feng, Y.; Han, S.; Dong, Q.; Cui, M.; Tieu, K. Microglial exosomes facilitate alpha-synuclein transmission in Parkinson’s disease. Brain 2020, 143, 1476–1497. [Google Scholar] [CrossRef]
  179. Dilsizoglu Senol, A.; Samarani, M.; Syan, S.; Guardia, C.M.; Nonaka, T.; Liv, N.; Latour-Lambert, P.; Hasegawa, M.; Klumperman, J.; Bonifacino, J.S.; et al. alpha-Synuclein fibrils subvert lysosome structure and function for the propagation of protein misfolding between cells through tunneling nanotubes. PLoS Biol. 2021, 19, e3001287. [Google Scholar] [CrossRef]
  180. Stykel, M.G.; Humphries, K.M.; Kamski-Hennekam, E.; Buchner-Duby, B.; Porte-Trachsel, N.; Ryan, T.; Coackley, C.L.; Bamm, V.V.; Harauz, G.; Ryan, S.D. Alpha-Synuclein mutation impairs processing of endomembrane compartments and promotes exocytosis and seeding of alpha-synuclein pathology. Cell Rep. 2021, 35, 109099. [Google Scholar] [CrossRef]
  181. Si, X.L.; Fang, Y.J.; Li, L.F.; Gu, L.Y.; Yin, X.Z.; Jun, T.; Yan, Y.P.; Pu, J.L.; Zhang, B.R. From inflammasome to Parkinson’s disease: Does the NLRP3 inflammasome facilitate exosome secretion and exosomal alpha-synuclein transmission in Parkinson’s disease? Exp. Neurol. 2021, 336, 113525. [Google Scholar] [CrossRef]
  182. Zhao, Z.H.; Chen, Z.T.; Zhou, R.L.; Zhang, X.; Ye, Q.Y.; Wang, Y.Z. Increased DJ-1 and alpha-Synuclein in Plasma Neural-Derived Exosomes as Potential Markers for Parkinson’s Disease. Front. Aging Neurosci. 2018, 10, 438. [Google Scholar] [CrossRef]
  183. Niu, M.; Li, Y.; Li, G.; Zhou, L.; Luo, N.; Yao, M.; Kang, W.; Liu, J. A longitudinal study on alpha-synuclein in plasma neuronal exosomes as a biomarker for Parkinson’s disease development and progression. Eur. J. Neurol. 2020, 27, 967–974. [Google Scholar] [CrossRef]
  184. Xia, Y.; Zhang, G.; Han, C.; Ma, K.; Guo, X.; Wan, F.; Kou, L.; Yin, S.; Liu, L.; Huang, J.; et al. Microglia as modulators of exosomal alpha-synuclein transmission. Cell Death Dis. 2019, 10, 174. [Google Scholar] [CrossRef] [Green Version]
  185. Jiang, Y.; Liu, J.; Chen, L.; Jin, Y.; Zhang, G.; Lin, Z.; Du, S.; Fu, Z.; Chen, T.; Qin, Y.; et al. Serum secreted miR-137-containing exosomes affects oxidative stress of neurons by regulating OXR1 in Parkinson’s disease. Brain Res. 2019, 1722, 146331. [Google Scholar] [CrossRef]
  186. Li, N.; Pan, X.; Zhang, J.; Ma, A.; Yang, S.; Ma, J.; Xie, A. Plasma levels of miR-137 and miR-124 are associated with Parkinson’s disease but not with Parkinson’s disease with depression. Neurol. Sci. 2017, 38, 761–767. [Google Scholar] [CrossRef]
  187. Li, Q.; Wang, Z.; Xing, H.; Wang, Y.; Guo, Y. Exosomes derived from miR-188-3p-modified adipose-derived mesenchymal stem cells protect Parkinson’s disease. Mol. Ther. Nucleic Acids 2021, 23, 1334–1344. [Google Scholar] [CrossRef]
  188. Sun, T.; Ding, Z.X.; Luo, X.; Liu, Q.S.; Cheng, Y. Blood Exosomes Have Neuroprotective Effects in a Mouse Model of Parkinson’s Disease. Oxidative Med. Cell. Longev. 2020, 2020, 3807476. [Google Scholar] [CrossRef]
  189. Rani, K.; Mukherjee, R.; Singh, E.; Kumar, S.; Sharma, V.; Vishwakarma, P.; Bharti, P.S.; Nikolajeff, F.; Dinda, A.K.; Goyal, V.; et al. Neuronal exosomes in saliva of Parkinson’s disease patients: A pilot study. Park. Relat. Disord. 2019, 67, 21–23. [Google Scholar] [CrossRef]
  190. Nonaka, T.; Wong, D.T.W. Liquid Biopsy in Head and Neck Cancer: Promises and Challenges. J. Dent. Res. 2018, 97, 701–708. [Google Scholar] [CrossRef]
  191. Theodoraki, M.N.; Ludwig, S. Exosomen: Potenzielle Flussigbiopsie bei Kopf-Hals-Karzinomen. [Exosomes: Potential liquid biopsy in head and neck cancer]. HNO 2020, 68, 106–110. [Google Scholar] [CrossRef]
  192. Mouliere, F.; Thierry, A.R. The importance of examining the proportion of circulating DNA originating from tumor, microenvironment and normal cells in colorectal cancer patients. Expert Opin. Biol. Ther. 2012, 12 (Suppl. 1), S209–S215. [Google Scholar] [CrossRef] [PubMed]
  193. Phallen, J.; Sausen, M.; Adleff, V.; Leal, A.; Hruban, C.; White, J.; Anagnostou, V.; Fiksel, J.; Cristiano, S.; Papp, E.; et al. Direct detection of early-stage cancers using circulating tumor DNA. Sci. Transl. Med. 2017, 9, eaan2415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Thakur, B.K.; Zhang, H.; Becker, A.; Matei, I.; Huang, Y.; Costa-Silva, B.; Zheng, Y.; Hoshino, A.; Brazier, H.; Xiang, J.; et al. Double-stranded DNA in exosomes: A novel biomarker in cancer detection. Cell Res. 2014, 24, 766–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Warnakulasuriya, S. Global epidemiology of oral and oropharyngeal cancer. Oral Oncol. 2009, 45, 309–316. [Google Scholar] [CrossRef] [PubMed]
  196. Georgaki, M.; Theofilou, V.I.; Pettas, E.; Stoufi, E.; Younis, R.H.; Kolokotronis, A.; Sauk, J.J.; Nikitakis, N.G. Understanding the complex pathogenesis of oral cancer: A comprehensive review. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2021, 132, 566–579. [Google Scholar] [CrossRef]
  197. Ram, H.; Sarkar, J.; Kumar, H.; Konwar, R.; Bhatt, M.L.; Mohammad, S. Oral cancer: Risk factors and molecular pathogenesis. J. Maxillofac. Oral Surg. 2011, 10, 132–137. [Google Scholar] [CrossRef] [Green Version]
  198. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Primers 2020, 6, 92. [Google Scholar] [CrossRef]
  199. Sarode, G.; Maniyar, N.; Sarode, S.C.; Jafer, M.; Patil, S.; Awan, K.H. Epidemiologic aspects of oral cancer. Dis. Mon. 2020, 66, 100988. [Google Scholar] [CrossRef]
  200. Sciubba, J.J. Oral cancer. The importance of early diagnosis and treatment. Am. J. Clin. Dermatol. 2001, 2, 239–251. [Google Scholar] [CrossRef]
  201. Westgaard, K.L.; Hynne, H.; Amdal, C.D.; Young, A.; Singh, P.B.; Chen, X.; Rykke, M.; Hove, L.H.; Aqrawi, L.A.; Utheim, T.P.; et al. Oral and ocular late effects in head and neck cancer patients treated with radiotherapy. Sci. Rep. 2021, 11, 4026. [Google Scholar] [CrossRef]
  202. Marioni, G.; Doro, D.; Marino, F.; Verdecchia, P.; Staffieri, C.; Staffieri, A. Skin and eye: Uncommon sites of distant metastasis from tongue base squamous cell carcinoma. Acta Otolaryngol. 2003, 123, 1110–1114. [Google Scholar] [CrossRef]
  203. Binkley, E.M.; Sampson, A.D.; Syed, N.A.; Boldt, H.C. Metastatic Squamous Cell Carcinoma of the Tonsil Mimicking Choroidal Melanoma. Ocul. Oncol. Pathol. 2020, 6, 405–409. [Google Scholar] [CrossRef] [PubMed]
  204. Xie, C.; Du, L.Y.; Guo, F.; Li, X.; Cheng, B. Exosomes derived from microRNA-101-3p-overexpressing human bone marrow mesenchymal stem cells suppress oral cancer cell proliferation, invasion, and migration. Mol. Cell. Biochem. 2019, 458, 11–26. [Google Scholar] [CrossRef]
  205. Liu, M.X.; Liao, J.; Xie, M.; Gao, Z.K.; Wang, X.H.; Zhang, Y.; Shang, M.H.; Yin, L.H.; Pu, Y.P.; Liu, R. miR-93-5p Transferred by Exosomes Promotes the Proliferation of Esophageal Cancer Cells via Intercellular Communication by Targeting PTEN. Biomed. Environ. Sci. 2018, 31, 171–185. [Google Scholar] [CrossRef]
  206. Ludwig, S.; Floros, T.; Theodoraki, M.N.; Hong, C.S.; Jackson, E.K.; Lang, S.; Whiteside, T.L. Suppression of Lymphocyte Functions by Plasma Exosomes Correlates with Disease Activity in Patients with Head and Neck Cancer. Clin. Cancer Res. 2017, 23, 4843–4854. [Google Scholar] [CrossRef] [Green Version]
  207. Langevin, S.; Kuhnell, D.; Parry, T.; Biesiada, J.; Huang, S.; Wise-Draper, T.; Casper, K.; Zhang, X.; Medvedovic, M.; Kasper, S. Comprehensive microRNA-sequencing of exosomes derived from head and neck carcinoma cells in vitro reveals common secretion profiles and potential utility as salivary biomarkers. Oncotarget 2017, 8, 82459–82474. [Google Scholar] [CrossRef] [Green Version]
  208. Wang, Y.; Springer, S.; Mulvey, C.L.; Silliman, N.; Schaefer, J.; Sausen, M.; James, N.; Rettig, E.M.; Guo, T.; Pickering, C.R.; et al. Detection of somatic mutations and HPV in the saliva and plasma of patients with head and neck squamous cell carcinomas. Sci. Transl. Med. 2015, 7, 293ra104. [Google Scholar] [CrossRef] [Green Version]
  209. Gai, C.; Camussi, F.; Broccoletti, R.; Gambino, A.; Cabras, M.; Molinaro, L.; Carossa, S.; Camussi, G.; Arduino, P.G. Salivary extracellular vesicle-associated miRNAs as potential biomarkers in oral squamous cell carcinoma. BMC Cancer 2018, 18, 439. [Google Scholar] [CrossRef] [Green Version]
  210. Lin, Y.; Dong, H.; Deng, W.; Lin, W.; Li, K.; Xiong, X.; Guo, Y.; Zhou, F.; Ma, C.; Chen, Y.; et al. Evaluation of Salivary Exosomal Chimeric GOLM1-NAA35 RNA as a Potential Biomarker in Esophageal Carcinoma. Clin. Cancer Res. 2019, 25, 3035–3045. [Google Scholar] [CrossRef]
  211. Cai, J.; Qiao, B.; Gao, N.; Lin, N.; He, W. Oral squamous cell carcinoma-derived exosomes promote M2 subtype macrophage polarization mediated by exosome-enclosed miR-29a-3p. Am. J. Physiol. Cell Physiol. 2019, 316, C731–C740. [Google Scholar] [CrossRef]
  212. Liu, C.J.; Lin, S.C.; Yang, C.C.; Cheng, H.W.; Chang, K.W. Exploiting salivary miR-31 as a clinical biomarker of oral squamous cell carcinoma. Head Neck 2012, 34, 219–224. [Google Scholar] [CrossRef] [PubMed]
  213. Ferlay, J.; Colombet, M.; Soerjomataram, I.; Parkin, D.M.; Pineros, M.; Znaor, A.; Bray, F. Cancer statistics for the year 2020: An overview. Int. J. Cancer 2021, 149, 778–789. [Google Scholar] [CrossRef] [PubMed]
  214. Lei, S.; Zheng, R.; Zhang, S.; Wang, S.; Chen, R.; Sun, K.; Zeng, H.; Zhou, J.; Wei, W. Global patterns of breast cancer incidence and mortality: A population-based cancer registry data analysis from 2000 to 2020. Cancer Commun. 2021, 41, 1183–1194. [Google Scholar] [CrossRef] [PubMed]
  215. Wilkinson, L.; Gathani, T. Understanding breast cancer as a global health concern. Br. J. Radiol. 2022, 95, 20211033. [Google Scholar] [CrossRef]
  216. Freedman, M.I.; Folk, J.C. Metastatic tumors to the eye and orbit. Patient survival and clinical characteristics. Arch. Ophthalmol. 1987, 105, 1215–1219. [Google Scholar] [CrossRef]
  217. Weiss, L. Comments on hematogenous metastatic patterns in humans as revealed by autopsy. Clin. Exp. Metastasis 1992, 10, 191–199. [Google Scholar] [CrossRef]
  218. McCartney, A. Intraocular metastasis. Br. J. Ophthalmol. 1993, 77, 133. [Google Scholar] [CrossRef] [Green Version]
  219. Konstantinidis, L.; Rospond-Kubiak, I.; Zeolite, I.; Heimann, H.; Groenewald, C.; Coupland, S.E.; Damato, B. Management of patients with uveal metastases at the Liverpool Ocular Oncology Centre. Br. J. Ophthalmol. 2014, 98, 92–98. [Google Scholar] [CrossRef]
  220. Kanthan, G.L.; Jayamohan, J.; Yip, D.; Conway, R.M. Management of metastatic carcinoma of the uveal tract: An evidence-based analysis. Clin. Exp. Ophthalmol. 2007, 35, 553–565. [Google Scholar] [CrossRef]
  221. Shtam, T.; Naryzhny, S.; Samsonov, R.; Karasik, D.; Mizgirev, I.; Kopylov, A.; Petrenko, E.; Zabrodskaya, Y.; Kamyshinsky, R.; Nikitin, D.; et al. Plasma exosomes stimulate breast cancer metastasis through surface interactions and activation of FAK signaling. Breast Cancer Res. Treat. 2019, 174, 129–141. [Google Scholar] [CrossRef]
  222. Shojaei, S.; Hashemi, S.M.; Ghanbarian, H.; Sharifi, K.; Salehi, M.; Mohammadi-Yeganeh, S. Delivery of miR-381-3p Mimic by Mesenchymal Stem Cell-Derived Exosomes Inhibits Triple Negative Breast Cancer Aggressiveness; an In Vitro Study. Stem Cell Rev. Rep. 2021, 17, 1027–1038. [Google Scholar] [CrossRef]
  223. Sun, L.; He, M.; Xu, N.; Xu, D.H.; Ben-David, Y.; Yang, Z.Y.; Li, Y.J. Regulation of RAB22A by mir-193b inhibits breast cancer growth and metastasis mediated by exosomes. Int. J. Oncol. 2018, 53, 2705–2714. [Google Scholar] [CrossRef] [Green Version]
  224. Inubushi, S.; Kawaguchi, H.; Mizumoto, S.; Kunihisa, T.; Baba, M.; Kitayama, Y.; Takeuchi, T.; Hoffman, R.M.; Tanino, H.; Sasaki, R. Oncogenic miRNAs Identified in Tear Exosomes From Metastatic Breast Cancer Patients. Anticancer Res. 2020, 40, 3091–3096. [Google Scholar] [CrossRef]
  225. Ando, W.; Kikuchi, K.; Uematsu, T.; Yokomori, H.; Takaki, T.; Sogabe, M.; Kohgo, Y.; Otori, K.; Ishikawa, S.; Okazaki, I. Novel breast cancer screening: Combined expression of miR-21 and MMP-1 in urinary exosomes detects 95% of breast cancer without metastasis. Sci. Rep. 2019, 9, 13595. [Google Scholar] [CrossRef] [Green Version]
  226. Pakravan, K.; Babashah, S.; Sadeghizadeh, M.; Mowla, S.J.; Mossahebi-Mohammadi, M.; Ataei, F.; Dana, N.; Javan, M. MicroRNA-100 shuttled by mesenchymal stem cell-derived exosomes suppresses in vitro angiogenesis through modulating the mTOR/HIF-1alpha/VEGF signaling axis in breast cancer cells. Cell. Oncol. 2017, 40, 457–470. [Google Scholar] [CrossRef]
  227. Lee, J.K.; Park, S.R.; Jung, B.K.; Jeon, Y.K.; Lee, Y.S.; Kim, M.K.; Kim, Y.G.; Jang, J.Y.; Kim, C.W. Exosomes derived from mesenchymal stem cells suppress angiogenesis by down-regulating VEGF expression in breast cancer cells. PLoS ONE 2013, 8, e84256. [Google Scholar] [CrossRef] [Green Version]
  228. Yuan, L.; Liu, Y.; Qu, Y.; Liu, L.; Li, H. Exosomes Derived From MicroRNA-148b-3p-Overexpressing Human Umbilical Cord Mesenchymal Stem Cells Restrain Breast Cancer Progression. Front. Oncol. 2019, 9, 1076. [Google Scholar] [CrossRef] [Green Version]
  229. Mohd Ali, N.; Yeap, S.K.; Ho, W.Y.; Boo, L.; Ky, H.; Satharasinghe, D.A.; Tan, S.W.; Cheong, S.K.; Huang, H.D.; Lan, K.C.; et al. Adipose MSCs Suppress MCF7 and MDA-MB-231 Breast Cancer Metastasis and EMT Pathways Leading to Dormancy via Exosomal-miRNAs Following Co-Culture Interaction. Pharmaceuticals 2020, 14, 8. [Google Scholar] [CrossRef]
  230. Huang, X.; Zhang, H.; Guo, X.; Zhu, Z.; Cai, H.; Kong, X. Insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) in cancer. J. Hematol. Oncol. 2018, 11, 88. [Google Scholar] [CrossRef] [Green Version]
  231. Lau, C.S.; Wong, D.T. Breast cancer exosome-like microvesicles and salivary gland cells interplay alters salivary gland cell-derived exosome-like microvesicles in vitro. PLoS ONE 2012, 7, e33037. [Google Scholar] [CrossRef]
  232. Zhang, L.; Xiao, H.; Karlan, S.; Zhou, H.; Gross, J.; Elashoff, D.; Akin, D.; Yan, X.; Chia, D.; Karlan, B.; et al. Discovery and preclinical validation of salivary transcriptomic and proteomic biomarkers for the non-invasive detection of breast cancer. PLoS ONE 2010, 5, e15573. [Google Scholar] [CrossRef] [PubMed]
  233. Streckfus, C.F.; Mayorga-Wark, O.; Arreola, D.; Edwards, C.; Bigler, L.; Dubinsky, W.P. Breast cancer related proteins are present in saliva and are modulated secondary to ductal carcinoma in situ of the breast. Cancer Investig. 2008, 26, 159–167. [Google Scholar] [CrossRef]
  234. Zamanova, S.; Shabana, A.M.; Mondal, U.K.; Ilies, M.A. Carbonic anhydrases as disease markers. Expert Opin. Ther. Pat. 2019, 29, 509–533. [Google Scholar] [CrossRef] [PubMed]
  235. Rho, S.B.; Lee, J.H.; Park, M.S.; Byun, H.J.; Kang, S.; Seo, S.S.; Kim, J.Y.; Park, S.Y. Anti-apoptotic protein TCTP controls the stability of the tumor suppressor p53. FEBS Lett. 2011, 585, 29–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  237. Cruzado-Sanchez, D.; Saavedra-Mejia, L.A.; Tellez, W.A.; Maquera-Torres, G.; Serpa-Frias, S. Metastatic Intraocular Tumor Due to Colorectal Adenocarcinoma: Case Report and Literature Review. J. Ophthalmic Vis. Res. 2020, 15, 565–570. [Google Scholar] [CrossRef]
  238. Khawaja, M.R.; Minturn, J.T.; Spittler, A.J.; Chiorean, E.G. Ocular metastasis of colorectal cancer: An uncommon presentation of a common malignancy. Hematol. Oncol. Stem Cell Ther. 2015, 8, 176–180. [Google Scholar] [CrossRef] [Green Version]
  239. Jiang, K.; Chen, H.; Fang, Y.; Chen, L.; Zhong, C.; Bu, T.; Dai, S.; Pan, X.; Fu, D.; Qian, Y.; et al. Exosomal ANGPTL1 attenuates colorectal cancer liver metastasis by regulating Kupffer cell secretion pattern and impeding MMP9 induced vascular leakiness. J. Exp. Clin. Cancer Res. 2021, 40, 21. [Google Scholar] [CrossRef]
  240. Xiao, Y.; Li, Y.; Yuan, Y.; Liu, B.; Pan, S.; Liu, Q.; Qi, X.; Zhou, H.; Dong, W.; Jia, L. The potential of exosomes derived from colorectal cancer as a biomarker. Clin. Chim. Acta 2019, 490, 186–193. [Google Scholar] [CrossRef]
  241. Li, Y.; Zheng, Q.; Bao, C.; Li, S.; Guo, W.; Zhao, J.; Chen, D.; Gu, J.; He, X.; Huang, S. Circular RNA is enriched and stable in exosomes: A promising biomarker for cancer diagnosis. Cell Res. 2015, 25, 981–984. [Google Scholar] [CrossRef] [Green Version]
  242. Sazanov, A.A.; Kiselyova, E.V.; Zakharenko, A.A.; Romanov, M.N.; Zaraysky, M.I. Plasma and saliva miR-21 expression in colorectal cancer patients. J. Appl. Genet. 2017, 58, 231–237. [Google Scholar] [CrossRef]
  243. Rapado-Gonzalez, O.; Majem, B.; Alvarez-Castro, A.; Diaz-Pena, R.; Abalo, A.; Suarez-Cabrera, L.; Gil-Moreno, A.; Santamaria, A.; Lopez-Lopez, R.; Muinelo-Romay, L.; et al. A Novel Saliva-Based miRNA Signature for Colorectal Cancer Diagnosis. J. Clin. Med. 2019, 8, 2029. [Google Scholar] [CrossRef] [Green Version]
  244. Guo, Y.; Wang, X.; Xiao, J.; Xu, Y.; Cai, Y.; Sun, C.; Ma, K. Lung squamous cell carcinoma with solitary ocular metastasis and its successful treatment with thoracic surgery and chemotherapy: An interesting and rare case report. BMC Cancer 2018, 18, 1004. [Google Scholar] [CrossRef] [Green Version]
  245. Qu, Z.; Liu, J.; Zhu, L.; Zhou, Q. A Comprehensive Understanding of Choroidal Metastasis from Lung Cancer. Onco Targets Ther. 2021, 14, 4451–4465. [Google Scholar] [CrossRef]
  246. Chen, H.F.; Wang, W.X.; Li, X.F.; Wu, L.X.; Zhu, Y.C.; Du, K.Q.; Xu, C.W. Eye metastasis in lung adenocarcinoma mimicking anterior scleritis: A case report. World J. Clin. Cases 2020, 8, 410–414. [Google Scholar] [CrossRef]
  247. Lampaki, S.; Kioumis, I.; Pitsiou, G.; Lazaridis, G.; Syrigos, K.; Trakada, G.; Kakolyris, S.; Zarogoulidis, K.; Mpoukovinas, I.; Rapti, A.; et al. Lung cancer and eye metastases. Med. Hypothesis Discov. Innov. Ophthalmol. 2014, 3, 40–44. [Google Scholar]
  248. Liu, S.L.; Nie, Y.H.; He, T.; Yan, X.X.; Xing, Y.Q. Iris metastasis as the first sign of small cell lung cancer: A case report. Oncol. Lett. 2017, 13, 1547–1552. [Google Scholar] [CrossRef] [Green Version]
  249. Sakellakis, M.; Peroukides, S.; Iconomou, G.; Kalofonos, H. Iris Metastasis in a Patient With Small Cell Lung Cancer: A Case Report. Iran. Red. Crescent Med. J. 2016, 18, e21522. [Google Scholar] [CrossRef] [Green Version]
  250. Wong, M.; Frank, J.H.; Shields, C.L. Non-small cell lung cancer with iris metastasis controlled with osimertinib and monthly intravitreal bevacizumab. Am. J. Ophthalmol. Case Rep. 2022, 25, 101269. [Google Scholar] [CrossRef]
  251. Rahman, M.A.; Barger, J.F.; Lovat, F.; Gao, M.; Otterson, G.A.; Nana-Sinkam, P. Lung cancer exosomes as drivers of epithelial mesenchymal transition. Oncotarget 2016, 7, 54852–54866. [Google Scholar] [CrossRef]
  252. Xiao, X.; Yu, S.; Li, S.; Wu, J.; Ma, R.; Cao, H.; Zhu, Y.; Feng, J. Exosomes: Decreased sensitivity of lung cancer A549 cells to cisplatin. PLoS ONE 2014, 9, e89534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  253. Zhang, Y.; Liu, Z.; Li, S.; Wang, M.; Dai, D.; Jing, H.; Liu, L. Upregulation of E-cadherin in bronchoalveolar lavage fluid-derived exosomes in patients with lung cancer. Thorac. Cancer 2020, 11, 41–47. [Google Scholar] [CrossRef] [PubMed]
  254. Wang, H.; Wang, L.; Pan, H.; Wang, Y.; Shi, M.; Yu, H.; Wang, C.; Pan, X.; Chen, Z. Exosomes Derived From Macrophages Enhance Aerobic Glycolysis and Chemoresistance in Lung Cancer by Stabilizing c-Myc via the Inhibition of NEDD4L. Front. Cell Dev. Biol. 2020, 8, 620603. [Google Scholar] [CrossRef] [PubMed]
  255. Sun, Y.; Huo, C.; Qiao, Z.; Shang, Z.; Uzzaman, A.; Liu, S.; Jiang, X.; Fan, L.Y.; Ji, L.; Guan, X.; et al. Comparative Proteomic Analysis of Exosomes and Microvesicles in Human Saliva for Lung Cancer. J. Proteome Res. 2018, 17, 1101–1107. [Google Scholar] [CrossRef]
  256. Bian, B.; Zhao, C.; He, X.; Gong, Y.; Ren, C.; Ge, L.; Zeng, Y.; Li, Q.; Chen, M.; Weng, C.; et al. Exosomes derived from neural progenitor cells preserve photoreceptors during retinal degeneration by inactivating microglia. J. Extracell. Vesicles 2020, 9, 1748931. [Google Scholar] [CrossRef] [Green Version]
  257. Ragusa, M.; Barbagallo, C.; Statello, L.; Caltabiano, R.; Russo, A.; Puzzo, L.; Avitabile, T.; Longo, A.; Toro, M.D.; Barbagallo, D.; et al. miRNA profiling in vitreous humor, vitreal exosomes and serum from uveal melanoma patients: Pathological and diagnostic implications. Cancer Biol. Ther. 2015, 16, 1387–1396. [Google Scholar] [CrossRef]
  258. Dong, H.; Wang, M.; Li, Q. Exosomal miR-4488 and miR-1273g-5p inhibit the epithelial-mesenchymal transition of transforming growth factor beta2-mediated retinal pigment epithelial cells by targeting ATP-binding cassette A4. Bioengineered 2021, 12, 9693–9706. [Google Scholar] [CrossRef]
  259. Liu, H.; Yuan, W.; Pang, Q.; Xue, C.; Yan, X. Single-particle analysis of tear fluid reveals abundant presence of tissue factor-exposing extracellular vesicles with strong coagulation activity. Talanta 2022, 239, 123089. [Google Scholar] [CrossRef]
  260. Lee, R.; Wong, T.Y.; Sabanayagam, C. Epidemiology of diabetic retinopathy, diabetic macular edema and related vision loss. Eye Vis. 2015, 2, 17. [Google Scholar] [CrossRef] [Green Version]
  261. Wong, T.Y.; Sabanayagam, C. Strategies to Tackle the Global Burden of Diabetic Retinopathy: From Epidemiology to Artificial Intelligence. Ophthalmologica 2020, 243, 9–20. [Google Scholar] [CrossRef]
  262. Ogurtsova, K.; da Rocha Fernandes, J.D.; Huang, Y.; Linnenkamp, U.; Guariguata, L.; Cho, N.H.; Cavan, D.; Shaw, J.E.; Makaroff, L.E. IDF Diabetes Atlas: Global estimates for the prevalence of diabetes for 2015 and 2040. Diabetes Res. Clin. Pract. 2017, 128, 40–50. [Google Scholar] [CrossRef] [Green Version]
  263. Magliano, D.J.; Boyko, E.J. IDF Diabetes Atlas, 10th ed.; International Diabetes Federation: Brussels, Belgium, 2021. [Google Scholar]
  264. Wilkinson, C.P.; Ferris, F.L., 3rd; Klein, R.E.; Lee, P.P.; Agardh, C.D.; Davis, M.; Dills, D.; Kampik, A.; Pararajasegaram, R.; Verdaguer, J.T.; et al. Proposed international clinical diabetic retinopathy and diabetic macular edema disease severity scales. Ophthalmology 2003, 110, 1677–1682. [Google Scholar] [CrossRef]
  265. Aguilar, E.; Friedlander, M.; Gariano, R.F. Endothelial proliferation in diabetic retinal microaneurysms. Arch. Ophthalmol. 2003, 121, 740–741. [Google Scholar] [CrossRef]
  266. Cherian, S.; Roy, S.; Pinheiro, A.; Roy, S. Tight glycemic control regulates fibronectin expression and basement membrane thickening in retinal and glomerular capillaries of diabetic rats. Investig. Ophthalmol. Vis. Sci. 2009, 50, 943–949. [Google Scholar] [CrossRef] [Green Version]
  267. Lee, T.H.; Hsieh, S.T.; Chiang, H.Y. Fibronectin inhibitor pUR4 attenuates tumor necrosis factor alpha-induced endothelial hyperpermeability by modulating beta1 integrin activation. J. Biomed. Sci. 2019, 26, 37. [Google Scholar] [CrossRef] [Green Version]
  268. Wong, T.Y.; Cheung, C.M.; Larsen, M.; Sharma, S.; Simo, R. Diabetic retinopathy. Nat. Rev. Dis. Primers 2016, 2, 16012. [Google Scholar] [CrossRef]
  269. Newman, D.K. Surgical management of the late complications of proliferative diabetic retinopathy. Eye 2010, 24, 441–449. [Google Scholar] [CrossRef]
  270. Gunduz, K.; Bakri, S.J. Management of proliferative diabetic retinopathy. Compr. Ophthalmol. Update 2007, 8, 245–256. [Google Scholar]
  271. Li, D.Q.; Choudhry, N. Tractional Retinal Detachment Secondary to Diabetic Retinopathy. JAMA Ophthalmol. 2018, 136, e183507. [Google Scholar] [CrossRef]
  272. Maisto, R.; Trotta, M.C.; Petrillo, F.; Izzo, S.; Cuomo, G.; Alfano, R.; Hermenean, A.; Barcia, J.M.; Galdiero, M.; Platania, C.B.M.; et al. Resolvin D1 Modulates the Intracellular VEGF-Related miRNAs of Retinal Photoreceptors Challenged with High Glucose. Front. Pharmacol. 2020, 11, 235. [Google Scholar] [CrossRef] [Green Version]
  273. Tokarz, A.; Szuscik, I.; Kusnierz-Cabala, B.; Kapusta, M.; Konkolewska, M.; Zurakowski, A.; Georgescu, A.; Stepien, E. Extracellular vesicles participate in the transport of cytokines and angiogenic factors in diabetic patients with ocular complications. Folia Med. Crac. 2015, 55, 35–48. [Google Scholar]
  274. Huang, C.; Fisher, K.P.; Hammer, S.S.; Busik, J.V. Extracellular Vesicle-Induced Classical Complement Activation Leads to Retinal Endothelial Cell Damage via MAC Deposition. Int. J. Mol. Sci. 2020, 21, 1693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  275. 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. Exp. Eye Res. 2019, 189, 107813. [Google Scholar] [CrossRef] [PubMed]
  276. Kamalden, T.A.; Macgregor-Das, A.M.; Kannan, S.M.; Dunkerly-Eyring, B.; Khaliddin, N.; Xu, Z.; Fusco, A.P.; Yazib, S.A.; Chow, R.C.; Duh, E.J.; et al. Exosomal MicroRNA-15a Transfer from the Pancreas Augments Diabetic Complications by Inducing Oxidative Stress. Antioxid. Redox Signal. 2017, 27, 913–930. [Google Scholar] [CrossRef]
  277. Katome, T.; Namekata, K.; Mitamura, Y.; Semba, K.; Egawa, M.; Naito, T.; Harada, C.; Harada, T. Expression of intraocular peroxisome proliferator-activated receptor gamma in patients with proliferative diabetic retinopathy. J. Diabetes Complicat. 2015, 29, 275–281. [Google Scholar] [CrossRef]
  278. Huang, C.; Fisher, K.P.; Hammer, S.S.; Navitskaya, S.; Blanchard, G.J.; Busik, J.V. Plasma Exosomes Contribute to Microvascular Damage in Diabetic Retinopathy by Activating the Classical Complement Pathway. Diabetes 2018, 67, 1639–1649. [Google Scholar] [CrossRef] [Green Version]
  279. Wooff, Y.; Cioanca, A.V.; Chu-Tan, J.A.; Aggio-Bruce, R.; Schumann, U.; Natoli, R. Small-Medium Extracellular Vesicles and Their miRNA Cargo in Retinal Health and Degeneration: Mediators of Homeostasis, and Vehicles for Targeted Gene Therapy. Front. Cell. Neurosci. 2020, 14, 160. [Google Scholar] [CrossRef]
  280. Gu, S.; Liu, Y.; Zou, J.; Wang, W.; Wei, T.; Wang, X.; Zhu, L.; Zhang, M.; Zhu, J.; Xie, T.; et al. Retinal pigment epithelial cells secrete miR-202-5p-containing exosomes to protect against proliferative diabetic retinopathy. Exp. Eye Res. 2020, 201, 108271. [Google Scholar] [CrossRef]
  281. Safwat, A.; Sabry, D.; Ragiae, A.; Amer, E.; Mahmoud, R.H.; Shamardan, R.M. Adipose mesenchymal stem cells-derived exosomes attenuate retina degeneration of streptozotocin-induced diabetes in rabbits. J. Circ. Biomark. 2018, 7, 1849454418807827. [Google Scholar] [CrossRef] [Green Version]
  282. Moisseiev, E.; Anderson, J.D.; Oltjen, S.; Goswami, M.; Zawadzki, R.J.; Nolta, J.A.; Park, S.S. Protective Effect of Intravitreal Administration of Exosomes Derived from Mesenchymal Stem Cells on Retinal Ischemia. Curr. Eye Res. 2017, 42, 1358–1367. [Google Scholar] [CrossRef] [Green Version]
  283. Liu, C.; Ge, H.M.; Liu, B.H.; Dong, R.; Shan, K.; Chen, X.; Yao, M.D.; Li, X.M.; Yao, J.; Zhou, R.M.; et al. Targeting pericyte-endothelial cell crosstalk by circular RNA-cPWWP2A inhibition aggravates diabetes-induced microvascular dysfunction. Proc. Natl. Acad. Sci. USA 2019, 116, 7455–7464. [Google Scholar] [CrossRef] [Green Version]
  284. Hamel, C. Retinitis pigmentosa. Orphanet J. Rare Dis. 2006, 1, 40. [Google Scholar] [CrossRef]
  285. Fishman, G.A. A historical perspective on the early treatment of night blindness and the use of dubious and unproven treatment strategies for patients with retinitis pigmentosa. Surv. Ophthalmol. 2013, 58, 652–663. [Google Scholar] [CrossRef]
  286. Sahaboglu, A.; Tanimoto, N.; Kaur, J.; Sancho-Pelluz, J.; Huber, G.; Fahl, E.; Arango-Gonzalez, B.; Zrenner, E.; Ekstrom, P.; Lowenheim, H.; et al. PARP1 gene knock-out increases resistance to retinal degeneration without affecting retinal function. PLoS ONE 2010, 5, e15495. [Google Scholar] [CrossRef] [Green Version]
  287. Arango-Gonzalez, B.; Trifunovic, D.; Sahaboglu, A.; Kranz, K.; Michalakis, S.; Farinelli, P.; Koch, S.; Koch, F.; Cottet, S.; Janssen-Bienhold, U.; et al. Identification of a common non-apoptotic cell death mechanism in hereditary retinal degeneration. PLoS ONE 2014, 9, e112142. [Google Scholar] [CrossRef]
  288. Vidal-Gil, L.; Sancho-Pelluz, J.; Zrenner, E.; Oltra, M.; Sahaboglu, A. Poly ADP ribosylation and extracellular vesicle activity in rod photoreceptor degeneration. Sci. Rep. 2019, 9, 3758. [Google Scholar] [CrossRef] [Green Version]
  289. Thomas, C.J.; Mirza, R.G.; Gill, M.K. Age-Related Macular Degeneration. Med. Clin. N. Am. 2021, 105, 473–491. [Google Scholar] [CrossRef]
  290. Wang, A.L.; Lukas, T.J.; Yuan, M.; Du, N.; Tso, M.O.; Neufeld, A.H. Autophagy and exosomes in the aged retinal pigment epithelium: Possible relevance to drusen formation and age-related macular degeneration. PLoS ONE 2009, 4, e4160. [Google Scholar] [CrossRef] [Green Version]
  291. Flores, R.; Carneiro, A.; Vieira, M.; Tenreiro, S.; Seabra, M.C. Age-Related Macular Degeneration: Pathophysiology, Management, and Future Perspectives. Ophthalmologica 2021, 244, 495–511. [Google Scholar] [CrossRef]
  292. Bowes Rickman, C.; Farsiu, S.; Toth, C.A.; Klingeborn, M. Dry age-related macular degeneration: Mechanisms, therapeutic targets, and imaging. Investig. Ophthalmol. Vis. Sci. 2013, 54, ORSF68–ORSF80. [Google Scholar] [CrossRef] [Green Version]
  293. Bird, A.C. Therapeutic targets in age-related macular disease. J. Clin. Investig. 2010, 120, 3033–3041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  294. Blasiak, J. Senescence in the pathogenesis of age-related macular degeneration. Cell. Mol. Life Sci. 2020, 77, 789–805. [Google Scholar] [CrossRef] [PubMed]
  295. Penn, J.S.; Madan, A.; Caldwell, R.B.; Bartoli, M.; Caldwell, R.W.; Hartnett, M.E. Vascular endothelial growth factor in eye disease. Prog. Retin. Eye Res. 2008, 27, 331–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  296. Shao, J.; Choudhary, M.M.; Schachat, A.P. Neovascular Age-Related Macular Degeneration. Dev. Ophthalmol. 2016, 55, 125–136. [Google Scholar] [CrossRef]
  297. Sivaprasad, S.; Chong, N.V. The complement system and age-related macular degeneration. Eye 2006, 20, 867–872. [Google Scholar] [CrossRef]
  298. Scholl, H.P.; Charbel Issa, P.; Walier, M.; Janzer, S.; Pollok-Kopp, B.; Borncke, F.; Fritsche, L.G.; Chong, N.V.; Fimmers, R.; Wienker, T.; et al. Systemic complement activation in age-related macular degeneration. PLoS ONE 2008, 3, e2593. [Google Scholar] [CrossRef]
  299. Kang, G.Y.; Bang, J.Y.; Choi, A.J.; Yoon, J.; Lee, W.C.; Choi, S.; Yoon, S.; Kim, H.C.; Baek, J.H.; Park, H.S.; et al. Exosomal proteins in the aqueous humor as novel biomarkers in patients with neovascular age-related macular degeneration. J. Proteome Res. 2014, 13, 581–595. [Google Scholar] [CrossRef]
  300. Atienzar-Aroca, S.; Flores-Bellver, M.; Serrano-Heras, G.; Martinez-Gil, N.; Barcia, J.M.; Aparicio, S.; Perez-Cremades, D.; Garcia-Verdugo, J.M.; Diaz-Llopis, M.; Romero, F.J.; et al. Oxidative stress in retinal pigment epithelium cells increases exosome secretion and promotes angiogenesis in endothelial cells. J. Cell. Mol. Med. 2016, 20, 1457–1466. [Google Scholar] [CrossRef]
  301. Tong, Y.; Zhou, Y.L.; Wang, Y.X.; Zhao, P.Q.; Wang, Z.Y. Retinal pigment epithelium cell-derived exosomes: Possible relevance to CNV in wet-age related macular degeneration. Med. Hypotheses 2016, 97, 98–101. [Google Scholar] [CrossRef]
  302. ElShelmani, H.; Wride, M.A.; Saad, T.; Rani, S.; Kelly, D.J.; Keegan, D. The Role of Deregulated MicroRNAs in Age-Related Macular Degeneration Pathology. Transl. Vis. Sci. Technol. 2021, 10, 12. [Google Scholar] [CrossRef]
  303. Chen, N.; Wang, J.; Hu, Y.; Cui, B.; Li, W.; Xu, G.; Liu, L.; Liu, S. MicroRNA-410 reduces the expression of vascular endothelial growth factor and inhibits oxygen-induced retinal neovascularization. PLoS ONE 2014, 9, e95665. [Google Scholar] [CrossRef]
  304. Wang, L.; Lee, A.Y.; Wigg, J.P.; Peshavariya, H.; Liu, P.; Zhang, H. miR-126 Regulation of Angiogenesis in Age-Related Macular Degeneration in CNV Mouse Model. Int. J. Mol. Sci. 2016, 17, 895. [Google Scholar] [CrossRef] [Green Version]
  305. Ye, L.; Peng, Y.; Mo, J.; Yao, Y. MiR-126 enhances VEGF expression in induced pluripotent stem cell-derived retinal neural stem cells by targeting spred-1. Int. J. Clin. Exp. Pathol. 2018, 11, 1023–1030. [Google Scholar]
  306. Mak, H.K.; Yung, J.S.Y.; Weinreb, R.N.; Ng, S.H.; Cao, X.; Ho, T.Y.C.; Ng, T.K.; Chu, W.K.; Yung, W.H.; Choy, K.W.; et al. MicroRNA-19a-PTEN Axis Is Involved in the Developmental Decline of Axon Regenerative Capacity in Retinal Ganglion Cells. Mol. Ther. Nucleic Acids 2020, 21, 251–263. [Google Scholar] [CrossRef]
  307. Hajrasouliha, A.R.; Jiang, G.; Lu, Q.; Lu, H.; Kaplan, H.J.; Zhang, H.G.; Shao, H. Exosomes from retinal astrocytes contain antiangiogenic components that inhibit laser-induced choroidal neovascularization. J. Biol. Chem. 2013, 288, 28058–28067. [Google Scholar] [CrossRef] [Green Version]
  308. Samaeekia, R.; Rabiee, B.; Putra, I.; Shen, X.; Park, Y.J.; Hematti, P.; Eslani, M.; Djalilian, A.R. Effect of Human Corneal Mesenchymal Stromal Cell-derived Exosomes on Corneal Epithelial Wound Healing. Investig. Ophthalmol. Vis. Sci. 2018, 59, 5194–5200. [Google Scholar] [CrossRef] [Green Version]
  309. Shojaati, G.; Khandaker, I.; Funderburgh, M.L.; Mann, M.M.; Basu, R.; Stolz, D.B.; Geary, M.L.; Dos Santos, A.; Deng, S.X.; Funderburgh, J.L. Mesenchymal Stem Cells Reduce Corneal Fibrosis and Inflammation via Extracellular Vesicle-Mediated Delivery of miRNA. Stem Cells Transl. Med. 2019, 8, 1192–1201. [Google Scholar] [CrossRef] [Green Version]
  310. Han, K.Y.; Tran, J.A.; Chang, J.H.; Azar, D.T.; Zieske, J.D. Potential role of corneal epithelial cell-derived exosomes in corneal wound healing and neovascularization. Sci. Rep. 2017, 7, 40548. [Google Scholar] [CrossRef] [Green Version]
  311. Han, K.Y.; Dugas-Ford, J.; Seiki, M.; Chang, J.H.; Azar, D.T. Evidence for the Involvement of MMP14 in MMP2 Processing and Recruitment in Exosomes of Corneal Fibroblasts. Investig. Ophthalmol. Vis. Sci. 2015, 56, 5323–5329. [Google Scholar] [CrossRef] [Green Version]
  312. Leszczynska, A.; Kulkarni, M.; Ljubimov, A.V.; Saghizadeh, M. Exosomes from normal and diabetic human corneolimbal keratocytes differentially regulate migration, proliferation and marker expression of limbal epithelial cells. Sci. Rep. 2018, 8, 15173. [Google Scholar] [CrossRef] [Green Version]
  313. McKay, T.B.; Karamichos, D.; Hutcheon, A.E.K.; Guo, X.; Zieske, J.D. Corneal Epithelial-Stromal Fibroblast Constructs to Study Cell-Cell Communication in Vitro. Bioengineering 2019, 6, 110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  314. Tao, H.; Chen, X.; Cao, H.; Zheng, L.; Li, Q.; Zhang, K.; Han, Z.; Han, Z.C.; Guo, Z.; Li, Z.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Corneal Wound Repair. Stem Cells Int. 2019, 2019, 5738510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  315. Shen, T.; Zheng, Q.Q.; Shen, J.; Li, Q.S.; Song, X.H.; Luo, H.B.; Hong, C.Y.; Yao, K. Effects of Adipose-derived Mesenchymal Stem Cell Exosomes on Corneal Stromal Fibroblast Viability and Extracellular Matrix Synthesis. Chin. Med. J. 2018, 131, 704–712. [Google Scholar] [CrossRef] [PubMed]
  316. Yu, C.; Chen, P.; Xu, J.; Liu, Y.; Li, H.; Wang, L.; Di, G. hADSCs derived extracellular vesicles inhibit NLRP3inflammasome activation and dry eye. Sci. Rep. 2020, 10, 14521. [Google Scholar] [CrossRef]
  317. Wang, S.; Hou, Y.; Li, X.; Song, Z.; Sun, B.; Li, X.; Zhang, H. Comparison of exosomes derived from induced pluripotent stem cells and mesenchymal stem cells as therapeutic nanoparticles for treatment of corneal epithelial defects. Aging 2020, 12, 19546–19562. [Google Scholar] [CrossRef]
  318. Escandon, P.; Liu, A.; Nicholas, S.E.; Khan, A.; Riaz, K.M.; Karamichos, D. Unravelling Novel Roles of Salivary Exosomes in the Regulation of Human Corneal Stromal Cell Migration and Wound Healing. Int. J. Mol. Sci. 2022, 23, 4330. [Google Scholar] [CrossRef]
  319. Leibowitz, J.A.; Woods, A.T.; Kesselman, M.M.; Mayi, B.S. Uveitis as a Predictor of Predisposition to Autoimmunity. Cureus 2020, 12, e7451. [Google Scholar] [CrossRef] [Green Version]
  320. Forrester, J.V.; Kuffova, L.; Dick, A.D. Autoimmunity, Autoinflammation, and Infection in Uveitis. Am. J. Ophthalmol. 2018, 189, 77–85. [Google Scholar] [CrossRef] [Green Version]
  321. Prete, M.; Dammacco, R.; Fatone, M.C.; Racanelli, V. Autoimmune uveitis: Clinical, pathogenetic, and therapeutic features. Clin. Exp. Med. 2016, 16, 125–136. [Google Scholar] [CrossRef]
  322. Knickelbein, J.E.; Liu, B.; Arakelyan, A.; Zicari, S.; Hannes, S.; Chen, P.; Li, Z.; Grivel, J.C.; Chaigne-Delalande, B.; Sen, H.N.; et al. Modulation of Immune Responses by Extracellular Vesicles From Retinal Pigment Epithelium. Investig. Ophthalmol. Vis. Sci. 2016, 57, 4101–4107. [Google Scholar] [CrossRef] [Green Version]
  323. Shigemoto-Kuroda, T.; Oh, J.Y.; Kim, D.K.; Jeong, H.J.; Park, S.Y.; Lee, H.J.; Park, J.W.; Kim, T.W.; An, S.Y.; Prockop, D.J.; et al. MSC-derived Extracellular Vesicles Attenuate Immune Responses in Two Autoimmune Murine Models: Type 1 Diabetes and Uveoretinitis. Stem Cell Rep. 2017, 8, 1214–1225. [Google Scholar] [CrossRef] [Green Version]
  324. Bai, L.; Shao, H.; Wang, H.; Zhang, Z.; Su, C.; Dong, L.; Yu, B.; Chen, X.; Li, X.; Zhang, X. Effects of Mesenchymal Stem Cell-Derived Exosomes on Experimental Autoimmune Uveitis. Sci. Rep. 2017, 7, 4323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  325. Kang, M.; Choi, J.K.; Jittayasothorn, Y.; Egwuagu, C.E. Interleukin 35-Producing Exosomes Suppress Neuroinflammation and Autoimmune Uveitis. Front. Immunol. 2020, 11, 1051. [Google Scholar] [CrossRef] [PubMed]
  326. Jiang, G.; Yun, J.; Kaplan, H.J.; Zhao, Y.; Sun, D.; Shao, H. Vaccination with circulating exosomes in autoimmune uveitis prevents recurrent intraocular inflammation. Clin. Exp. Ophthalmol. 2021, 49, 1069–1077. [Google Scholar] [CrossRef] [PubMed]
  327. Kaliki, S.; Shields, C.L. Uveal melanoma: Relatively rare but deadly cancer. Eye 2017, 31, 241–257. [Google Scholar] [CrossRef] [Green Version]
  328. Hu, D.N.; Simon, J.D.; Sarna, T. Role of ocular melanin in ophthalmic physiology and pathology. Photochem. Photobiol. 2008, 84, 639–644. [Google Scholar] [CrossRef]
  329. Materin, M.A.; Faries, M.; Kluger, H.M. Molecular alternations in uveal melanoma. Curr. Probl. Cancer 2011, 35, 211–224. [Google Scholar] [CrossRef]
  330. Krantz, B.A.; Dave, N.; Komatsubara, K.M.; Marr, B.P.; Carvajal, R.D. Uveal melanoma: Epidemiology, etiology, and treatment of primary disease. Clin. Ophthalmol. 2017, 11, 279–289. [Google Scholar] [CrossRef] [Green Version]
  331. Tsering, T.; Laskaris, A.; Abdouh, M.; Bustamante, P.; Parent, S.; Jin, E.; Ferrier, S.T.; Arena, G.; Burnier, J.V. Uveal Melanoma-Derived Extracellular Vesicles Display Transforming Potential and Carry Protein Cargo Involved in Metastatic Niche Preparation. Cancers 2020, 12, 2923. [Google Scholar] [CrossRef]
  332. Wroblewska, J.P.; Lach, M.S.; Kulcenty, K.; Galus, L.; Suchorska, W.M.; Rosel, D.; Brabek, J.; Marszalek, A. The Analysis of Inflammation-Related Proteins in a Cargo of Exosomes Derived from the Serum of Uveal Melanoma Patients Reveals Potential Biomarkers of Disease Progression. Cancers 2021, 13, 3334. [Google Scholar] [CrossRef]
  333. Eldh, M.; Olofsson Bagge, R.; Lasser, C.; Svanvik, J.; Sjostrand, M.; Mattsson, J.; Lindner, P.; Choi, D.S.; Gho, Y.S.; Lotvall, J. MicroRNA in exosomes isolated directly from the liver circulation in patients with metastatic uveal melanoma. BMC Cancer 2014, 14, 962. [Google Scholar] [CrossRef] [Green Version]
  334. Ray, A.; Gombos, D.S.; Vats, T.S. Retinoblastoma: An overview. Indian J. Pediatr. 2012, 79, 916–921. [Google Scholar] [CrossRef]
  335. Castro-Magdonel, B.E.; Orjuela, M.; Alvarez-Suarez, D.E.; Camacho, J.; Cabrera-Munoz, L.; Sadowinski-Pine, S.; Medina-Sanson, A.; Lara-Molina, C.; Garcia-Vega, D.; Vazquez, Y.; et al. Circulating miRNome detection analysis reveals 537 miRNAS in plasma, 625 in extracellular vesicles and a discriminant plasma signature of 19 miRNAs in children with retinoblastoma from which 14 are also detected in corresponding primary tumors. PLoS ONE 2020, 15, e0231394. [Google Scholar] [CrossRef] [Green Version]
  336. Ravishankar, H.; Mangani, A.S.; Phoebe Moses, G.L.; Mani, S.P.; Parameswaran, S.; Khetan, V.; Ganesan, S.; Krishnakumar, S. Serum exosomal miRNA as biomarkers for Retinoblastoma. Exp. Eye Res. 2020, 199, 108184. [Google Scholar] [CrossRef]
  337. Chen, S.; Chen, X.; Qiu, J.; Chen, P.; Han, X.; Wu, Y.; Zhuang, J.; Yang, M.; Wu, C.; Wu, N.; et al. Exosomes derived from retinoblastoma cells enhance tumour deterioration by infiltrating the microenvironment. Oncol. Rep. 2021, 45, 278–290. [Google Scholar] [CrossRef]
  338. Pastor, J.C.; Rojas, J.; Pastor-Idoate, S.; Di Lauro, S.; Gonzalez-Buendia, L.; Delgado-Tirado, S. Proliferative vitreoretinopathy: A new concept of disease pathogenesis and practical consequences. Prog. Retin. Eye Res. 2016, 51, 125–155. [Google Scholar] [CrossRef]
  339. Claes, C.; Lafeta, A.P. Proliferative vitreoretinopathy. Dev. Ophthalmol. 2014, 54, 188–195. [Google Scholar] [CrossRef]
  340. Pastor, J.C.; de la Rua, E.R.; Martin, F. Proliferative vitreoretinopathy: Risk factors and pathobiology. Prog. Retin. Eye Res. 2002, 21, 127–144. [Google Scholar] [CrossRef]
  341. Zhang, Y.; Wang, K.; Pan, J.; Yang, S.; Yao, H.; Li, M.; Li, H.; Lei, H.; Jin, H.; Wang, F. Exosomes mediate an epithelial-mesenchymal transition cascade in retinal pigment epithelial cells: Implications for proliferative vitreoretinopathy. J. Cell. Mol. Med. 2020, 24, 13324–13335. [Google Scholar] [CrossRef]
  342. Wang, M.; Li, Q.; Dong, H. Proteomic evidence that ABCA4 is vital for traumatic proliferative vitreoretinopathy formation and development. Exp. Eye Res. 2019, 181, 232–239. [Google Scholar] [CrossRef]
  343. Tham, Y.C.; Li, X.; Wong, T.Y.; Quigley, H.A.; Aung, T.; Cheng, C.Y. Global prevalence of glaucoma and projections of glaucoma burden through 2040: A systematic review and meta-analysis. Ophthalmology 2014, 121, 2081–2090. [Google Scholar] [CrossRef]
  344. Allison, K.; Patel, D.; Alabi, O. Epidemiology of Glaucoma: The Past, Present, and Predictions for the Future. Cureus 2020, 12, e11686. [Google Scholar] [CrossRef] [PubMed]
  345. Gupta, P.K.; Asrani, S.; Freedman, S.F.; El-Dairi, M.; Bhatti, M.T. Differentiating glaucomatous from non-glaucomatous optic nerve cupping by optical coherence tomography. Open Neurol. J. 2011, 5, 1–7. [Google Scholar] [CrossRef] [PubMed]
  346. Zhang, Y.X.; Huang, H.B.; Wei, S.H. Clinical characteristics of nonglaucomatous optic disc cupping. Exp. Ther. Med. 2014, 7, 995–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  347. Fard, M.A.; Moghimi, S.; Sahraian, A.; Ritch, R. Optic nerve head cupping in glaucomatous and non-glaucomatous optic neuropathy. Br. J. Ophthalmol. 2019, 103, 374–378. [Google Scholar] [CrossRef]
  348. Killer, H.E.; Pircher, A. What is the optimal glaucoma treatment: Reducing aqueous humour production or facilitating its outflow? Eye 2020, 34, 1719–1721. [Google Scholar] [CrossRef]
  349. Vranka, J.A.; Kelley, M.J.; Acott, T.S.; Keller, K.E. Extracellular matrix in the trabecular meshwork: Intraocular pressure regulation and dysregulation in glaucoma. Exp. Eye Res. 2015, 133, 112–125. [Google Scholar] [CrossRef] [Green Version]
  350. Hoffman, E.A.; Perkumas, K.M.; Highstrom, L.M.; Stamer, W.D. Regulation of myocilin-associated exosome release from human trabecular meshwork cells. Investig. Ophthalmol. Vis. Sci. 2009, 50, 1313–1318. [Google Scholar] [CrossRef] [Green Version]
  351. Lerner, N.; Schreiber-Avissar, S.; Beit-Yannai, E. Extracellular vesicle-mediated crosstalk between NPCE cells and TM cells result in modulation of Wnt signalling pathway and ECM remodelling. J. Cell. Mol. Med. 2020, 24, 4646–4658. [Google Scholar] [CrossRef]
  352. Lerner, N.; Avissar, S.; Beit-Yannai, E. Extracellular vesicles mediate signaling between the aqueous humor producing and draining cells in the ocular system. PLoS ONE 2017, 12, e0171153. [Google Scholar] [CrossRef] [Green Version]
  353. Stamer, W.D.; Hoffman, E.A.; Luther, J.M.; Hachey, D.L.; Schey, K.L. Protein profile of exosomes from trabecular meshwork cells. J. Proteomics 2011, 74, 796–804. [Google Scholar] [CrossRef] [Green Version]
  354. Dismuke, W.M.; Challa, P.; Navarro, I.; Stamer, W.D.; Liu, Y. Human aqueous humor exosomes. Exp. Eye Res. 2015, 132, 73–77. [Google Scholar] [CrossRef] [Green Version]
  355. Jayaram, H.; Phillips, J.I.; Lozano, D.C.; Choe, T.E.; Cepurna, W.O.; Johnson, E.C.; Morrison, J.C.; Gattey, D.M.; Saugstad, J.A.; Keller, K.E. Comparison of MicroRNA Expression in Aqueous Humor of Normal and Primary Open-Angle Glaucoma Patients Using PCR Arrays: A Pilot Study. Investig. Ophthalmol. Vis. Sci. 2017, 58, 2884–2890. [Google Scholar] [CrossRef] [Green Version]
  356. Liu, Y.; Bailey, J.C.; Helwa, I.; Dismuke, W.M.; Cai, J.; Drewry, M.; Brilliant, M.H.; Budenz, D.L.; Christen, W.G.; Chasman, D.I.; et al. A Common Variant in MIR182 Is Associated with Primary Open-Angle Glaucoma in the NEIGHBORHOOD Consortium. Investig. Ophthalmol. Vis. Sci. 2016, 57, 4528–4535. [Google Scholar] [CrossRef] [Green Version]
  357. Mead, B.; Ahmed, Z.; Tomarev, S. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles Promote Neuroprotection in a Genetic DBA/2J Mouse Model of Glaucoma. Investig. Ophthalmol. Vis. Sci. 2018, 59, 5473–5480. [Google Scholar] [CrossRef] [Green Version]
  358. Pan, D.; Chang, X.; Xu, M.; Zhang, M.; Zhang, S.; Wang, Y.; Luo, X.; Xu, J.; Yang, X.; Sun, X. UMSC-derived exosomes promote retinal ganglion cells survival in a rat model of optic nerve crush. J. Chem. Neuroanat. 2019, 96, 134–139. [Google Scholar] [CrossRef]
  359. Sugali, C.K.; Rayana, N.P.; Dai, J.; Peng, M.; Harris, S.L.; Webber, H.C.; Liu, S.; Dixon, S.G.; Parekh, P.H.; Martin, E.A.; et al. The Canonical Wnt Signaling Pathway Inhibits the Glucocorticoid Receptor Signaling Pathway in the Trabecular Meshwork. Am. J. Pathol. 2021, 191, 1020–1035. [Google Scholar] [CrossRef]
  360. Villarreal, G., Jr.; Chatterjee, A.; Oh, S.S.; Oh, D.J.; Kang, M.H.; Rhee, D.J. Canonical wnt signaling regulates extracellular matrix expression in the trabecular meshwork. Investig. Ophthalmol. Vis. Sci. 2014, 55, 7433–7440. [Google Scholar] [CrossRef] [Green Version]
  361. Gonzalez, J.M., Jr. Existence of the canonical Wnt signaling pathway in the human trabecular meshwork. Investig. Ophthalmol. Vis. Sci. 2012, 53, 6972. [Google Scholar] [CrossRef] [Green Version]
  362. Wang, W.H.; McNatt, L.G.; Pang, I.H.; Millar, J.C.; Hellberg, P.E.; Hellberg, M.H.; Steely, H.T.; Rubin, J.S.; Fingert, J.H.; Sheffield, V.C.; et al. Increased expression of the WNT antagonist sFRP-1 in glaucoma elevates intraocular pressure. J. Clin. Investig. 2008, 118, 1056–1064. [Google Scholar] [CrossRef] [Green Version]
  363. Tabak, S.; Feinshtein, V.; Schreiber-Avissar, S.; Beit-Yannai, E. Non-Pigmented Ciliary Epithelium-Derived Extracellular Vesicles Loaded with SMAD7 siRNA Attenuate Wnt Signaling in Trabecular Meshwork Cells In Vitro. Pharmaceuticals 2021, 14, 858. [Google Scholar] [CrossRef]
  364. McDonnell, F.; O’Brien, C.; Wallace, D. The role of epigenetics in the fibrotic processes associated with glaucoma. J. Ophthalmol. 2014, 2014, 750459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  365. Trelford, C.B.; Denstedt, J.T.; Armstrong, J.J.; Hutnik, C.M.L. The Pro-Fibrotic Behavior of Human Tenon’s Capsule Fibroblasts in Medically Treated Glaucoma Patients. Clin. Ophthalmol. 2020, 14, 1391–1402. [Google Scholar] [CrossRef] [PubMed]
  366. Aires, I.D.; Santiago, A.R. Microglial exosomes in retinal neuroinflammation: Focus in glaucoma. Neural Regen. Res. 2021, 16, 1801–1802. [Google Scholar] [CrossRef]
  367. Ran, N.; Gao, X.; Dong, X.; Li, J.; Lin, C.; Geng, M.; Yin, H. Effects of exosome-mediated delivery of myostatin propeptide on functional recovery of mdx mice. Biomaterials 2020, 236, 119826. [Google Scholar] [CrossRef] [PubMed]
  368. Guo, Q.; Jiang, C. Delivery strategies for macromolecular drugs in cancer therapy. Acta Pharm. Sin. B 2020, 10, 979–986. [Google Scholar] [CrossRef]
  369. Zheng, M.; Huang, M.; Ma, X.; Chen, H.; Gao, X. Harnessing Exosomes for the Development of Brain Drug Delivery Systems. Bioconjug. Chem. 2019, 30, 994–1005. [Google Scholar] [CrossRef]
  370. Kim, M.S.; Haney, M.J.; Zhao, Y.; Mahajan, V.; Deygen, I.; Klyachko, N.L.; Inskoe, E.; Piroyan, A.; Sokolsky, M.; Okolie, O.; et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 2016, 12, 655–664. [Google Scholar] [CrossRef] [Green Version]
  371. Kim, H.; Jang, H.; Cho, H.; Choi, J.; Hwang, K.Y.; Choi, Y.; Kim, S.H.; Yang, Y. Recent Advances in Exosome-Based Drug Delivery for Cancer Therapy. Cancers 2021, 13, 4435. [Google Scholar] [CrossRef]
  372. He, J.; Ren, W.; Wang, W.; Han, W.; Jiang, L.; Zhang, D.; Guo, M. Exosomal targeting and its potential clinical application. Drug Deliv. Transl. Res. 2021, 12, 2385–2402. [Google Scholar] [CrossRef]
  373. Mehryab, F.; Rabbani, S.; Shahhosseini, S.; Shekari, F.; Fatahi, Y.; Baharvand, H.; Haeri, A. Exosomes as a next-generation drug delivery system: An update on drug loading approaches, characterization, and clinical application challenges. Acta Biomater. 2020, 113, 42–62. [Google Scholar] [CrossRef]
  374. Chen, H.; Wang, L.; Zeng, X.; Schwarz, H.; Nanda, H.S.; Peng, X.; Zhou, Y. Exosomes, a New Star for Targeted Delivery. Front. Cell Dev. Biol. 2021, 9, 751079. [Google Scholar] [CrossRef]
  375. Salarpour, S.; Forootanfar, H.; Pournamdari, M.; Ahmadi-Zeidabadi, M.; Esmaeeli, M.; Pardakhty, A. Paclitaxel incorporated exosomes derived from glioblastoma cells: Comparative study of two loading techniques. Daru 2019, 27, 533–539. [Google Scholar] [CrossRef]
  376. 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. Nanomedicine 2018, 14, 195–204. [Google Scholar] [CrossRef]
  377. Han, Q.; Xie, Q.R.; Li, F.; Cheng, Y.; Wu, T.; Zhang, Y.; Lu, X.; Wong, A.S.T.; Sha, J.; Xia, W. Targeted inhibition of SIRT6 via engineered exosomes impairs tumorigenesis and metastasis in prostate cancer. Theranostics 2021, 11, 6526–6541. [Google Scholar] [CrossRef]
  378. Limoni, S.K.; Moghadam, M.F.; Moazzeni, S.M.; Gomari, H.; Salimi, F. Engineered Exosomes for Targeted Transfer of siRNA to HER2 Positive Breast Cancer Cells. Appl. Biochem. Biotechnol. 2019, 187, 352–364. [Google Scholar] [CrossRef]
  379. Kase, Y.; Uzawa, K.; Wagai, S.; Yoshimura, S.; Yamamoto, J.I.; Toeda, Y.; Okubo, M.; Eizuka, K.; Ando, T.; Nobuchi, T.; et al. Engineered exosomes delivering specific tumor-suppressive RNAi attenuate oral cancer progression. Sci. Rep. 2021, 11, 5897. [Google Scholar] [CrossRef]
  380. Sayyed, A.A.; Gondaliya, P.; Mali, M.; Pawar, A.; Bhat, P.; Khairnar, A.; Arya, N.; Kalia, K. MiR-155 Inhibitor-Laden Exosomes Reverse Resistance to Cisplatin in a 3D Tumor Spheroid and Xenograft Model of Oral Cancer. Mol. Pharm. 2021, 18, 3010–3025. [Google Scholar] [CrossRef]
  381. Cohen, O.; Betzer, O.; Elmaliach-Pnini, N.; Motiei, M.; Sadan, T.; Cohen-Berkman, M.; Dagan, O.; Popovtzer, A.; Yosepovich, A.; Barhom, H.; et al. ‘Golden’ exosomes as delivery vehicles to target tumors and overcome intratumoral barriers: In vivo tracking in a model for head and neck cancer. Biomater. Sci. 2021, 9, 2103–2114. [Google Scholar] [CrossRef]
  382. Sheykhhasan, M.; Kalhor, N.; Sheikholeslami, A.; Dolati, M.; Amini, E.; Fazaeli, H. Exosomes of Mesenchymal Stem Cells as a Proper Vehicle for Transfecting miR-145 into the Breast Cancer Cell Line and Its Effect on Metastasis. Biomed. Res. Int. 2021, 2021, 5516078. [Google Scholar] [CrossRef]
  383. Ogawa, Y.; Akimoto, Y.; Ikemoto, M.; Goto, Y.; Ishikawa, A.; Ohta, S.; Takase, Y.; Kawakami, H.; Tsujimoto, M.; Yanoshita, R. Stability of human salivary extracellular vesicles containing dipeptidyl peptidase IV under simulated gastrointestinal tract conditions. Biochem. Biophys. Rep. 2021, 27, 101034. [Google Scholar] [CrossRef] [PubMed]
  384. Luan, X.; Sansanaphongpricha, K.; Myers, I.; Chen, H.; Yuan, H.; Sun, D. Engineering exosomes as refined biological nanoplatforms for drug delivery. Acta Pharmacol. Sin. 2017, 38, 754–763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  385. Liang, Y.; Duan, L.; Lu, J.; Xia, J. Engineering exosomes for targeted drug delivery. Theranostics 2021, 11, 3183–3195. [Google Scholar] [CrossRef] [PubMed]
  386. Choi, H.; Yim, H.; Park, C.; Ahn, S.H.; Ahn, Y.; Lee, A.; Yang, H.; Choi, C. Targeted Delivery of Exosomes Armed with Anti-Cancer Therapeutics. Membranes 2022, 12, 85. [Google Scholar] [CrossRef]
  387. Shafiei, M.; Ansari, M.N.M.; Razak, S.I.A.; Khan, M.U.A. A Comprehensive Review on the Applications of Exosomes and Liposomes in Regenerative Medicine and Tissue Engineering. Polymers 2021, 13, 2529. [Google Scholar] [CrossRef]
Figure 1. (A) The formation of exosomes and effects on cell–cell communication. The early endosome is formed by inward budding of the endosomal membrane and then matures to form a multivesicular body (MVB) containing intraluminal vesicles (ILVs) or exosomes. The exosome is secreted upon fusion with the cell membrane. Exosomes release their contents by (a) direct fusion, (b) surface protein binding, or (c) endocytosis. (B) Composition of the exosome. The exosome is a vesicle that can carry DNA, mRNA, miRNA, proteins, and lipids, etc., to modulate function of the recipient target cell.
Figure 1. (A) The formation of exosomes and effects on cell–cell communication. The early endosome is formed by inward budding of the endosomal membrane and then matures to form a multivesicular body (MVB) containing intraluminal vesicles (ILVs) or exosomes. The exosome is secreted upon fusion with the cell membrane. Exosomes release their contents by (a) direct fusion, (b) surface protein binding, or (c) endocytosis. (B) Composition of the exosome. The exosome is a vesicle that can carry DNA, mRNA, miRNA, proteins, and lipids, etc., to modulate function of the recipient target cell.
Ijms 24 06363 g001
Figure 2. Discriminatory power of upregulated miRNAs hsa-miR-140-5p (ac), hsa-miR-146a-5p (df), hsa-miR-628-5p (gi), and the miRNA panel (jl), in salivary exosomes (SE) from periodontally healthy, gingivitis, and periodontitis patients by using receiver operating characteristics (ROC) curves and area under the curve, AUC. Note: Rows 1 and 2 show discriminatory power between periodontitis or gingivitis vs. periodontal health of whole saliva miRNA (a,d,g,j) and SE mRNA (b,e,h,k); and Row 3 (c,f,i,j) shows the discriminatory power of upregulated miRNAs in saliva and SEs between gingivitis and periodontitis patients [51].
Figure 2. Discriminatory power of upregulated miRNAs hsa-miR-140-5p (ac), hsa-miR-146a-5p (df), hsa-miR-628-5p (gi), and the miRNA panel (jl), in salivary exosomes (SE) from periodontally healthy, gingivitis, and periodontitis patients by using receiver operating characteristics (ROC) curves and area under the curve, AUC. Note: Rows 1 and 2 show discriminatory power between periodontitis or gingivitis vs. periodontal health of whole saliva miRNA (a,d,g,j) and SE mRNA (b,e,h,k); and Row 3 (c,f,i,j) shows the discriminatory power of upregulated miRNAs in saliva and SEs between gingivitis and periodontitis patients [51].
Ijms 24 06363 g002
Figure 3. Discrimination power of up-regulated LPS (a), global 5mC (b), m6dA (c), P. gingivalis (d), T. denticola (e), E. corrodens (f), and F. nucleatum (g) in saliva and salivary SEs from healthy, gingivitis, and periodontitis patients by using receiver operating characteristic (ROC) curves and area under the curve (AUC) [76].
Figure 3. Discrimination power of up-regulated LPS (a), global 5mC (b), m6dA (c), P. gingivalis (d), T. denticola (e), E. corrodens (f), and F. nucleatum (g) in saliva and salivary SEs from healthy, gingivitis, and periodontitis patients by using receiver operating characteristic (ROC) curves and area under the curve (AUC) [76].
Ijms 24 06363 g003
Figure 4. TEM of corneal epithelial–stromal interactions. (A) The presence of secreted collagen and extracellular vesicles were apparent between human corneal epithelial cells (hCECs) and human corneal fibroblasts (hCFs) (dashed box enlarged in right panel). (B) Extracellular vesicles were also present in between hCF cell populations (dashed box enlarged in right panel). Arrowheads = extracellular vesicles; * = mitochondria; † = vacuole; and ‡ = lysosome. Magnification = 12,000× [313].
Figure 4. TEM of corneal epithelial–stromal interactions. (A) The presence of secreted collagen and extracellular vesicles were apparent between human corneal epithelial cells (hCECs) and human corneal fibroblasts (hCFs) (dashed box enlarged in right panel). (B) Extracellular vesicles were also present in between hCF cell populations (dashed box enlarged in right panel). Arrowheads = extracellular vesicles; * = mitochondria; † = vacuole; and ‡ = lysosome. Magnification = 12,000× [313].
Ijms 24 06363 g004
Figure 5. TEM of corneal stromal interactions at high magnification. Secreted extracellular vesicles were identified within the stroma (red arrowheads). Large aggregates of extracellular vesicles (50–330 nm) were also found within the stroma near adjacent cells (black ellipse). Asterisks (*) denote deposited collagen fibrils. Magnification = 21,000× (left panel) and 31,000× (right panel) [313].
Figure 5. TEM of corneal stromal interactions at high magnification. Secreted extracellular vesicles were identified within the stroma (red arrowheads). Large aggregates of extracellular vesicles (50–330 nm) were also found within the stroma near adjacent cells (black ellipse). Asterisks (*) denote deposited collagen fibrils. Magnification = 21,000× (left panel) and 31,000× (right panel) [313].
Ijms 24 06363 g005
Figure 6. Expression of fibrotic markers in stromal constructs and epithelial–stromal co-cultures. Expression of collagen type III (Col 3) in (A) hCF control constructs, and (B) epithelial–stromal (hCEC/hCF) co-culture. High magnification of a single plane of focus in the (C) epithelial layer, and (D) stromal layer, show high expression of collagen type III by hCFs and little expression by hCECs. Expression of α-smooth muscle actin in the (E) epithelial layer (max projection), and (F1) stromal layer (max projection), of a co-culture show similar high expression by hCFs with low expression by hCECs. (F2) Expression of α-smooth muscle actin in hCF control. Imaged using a 40× objective lens. Scale bar = 10 μm (C,D), and 50 μm (E,F) [313].
Figure 6. Expression of fibrotic markers in stromal constructs and epithelial–stromal co-cultures. Expression of collagen type III (Col 3) in (A) hCF control constructs, and (B) epithelial–stromal (hCEC/hCF) co-culture. High magnification of a single plane of focus in the (C) epithelial layer, and (D) stromal layer, show high expression of collagen type III by hCFs and little expression by hCECs. Expression of α-smooth muscle actin in the (E) epithelial layer (max projection), and (F1) stromal layer (max projection), of a co-culture show similar high expression by hCFs with low expression by hCECs. (F2) Expression of α-smooth muscle actin in hCF control. Imaged using a 40× objective lens. Scale bar = 10 μm (C,D), and 50 μm (E,F) [313].
Ijms 24 06363 g006
Figure 7. Fibronectin (cFN) expression in hCF stromal constructs and epithelial–stromal co-cultures. (A) hCF construct only, showing no fibronectin staining. (B1) Epithelial–stromal (hCEC/hCF) co-culture showing the expression of fibronectin at the epithelial–stromal interface, similar to (B2) rat cornea 1 week post-keratectomy. (C) Max projection image of the hCEC layer showing little expression in the epithelial layer. (D) Max projection image of the hCF layer showing high fibronectin expression at the epithelial–stromal interface. Imaged using a 40× objective lens [313].
Figure 7. Fibronectin (cFN) expression in hCF stromal constructs and epithelial–stromal co-cultures. (A) hCF construct only, showing no fibronectin staining. (B1) Epithelial–stromal (hCEC/hCF) co-culture showing the expression of fibronectin at the epithelial–stromal interface, similar to (B2) rat cornea 1 week post-keratectomy. (C) Max projection image of the hCEC layer showing little expression in the epithelial layer. (D) Max projection image of the hCF layer showing high fibronectin expression at the epithelial–stromal interface. Imaged using a 40× objective lens [313].
Ijms 24 06363 g007
Figure 8. Localization of fibronectin (FN) in corneal epithelial–stromal co-cultures (hCE-TJ/hCF) at 3 days post-airlift. High expression of fibronectin in the epithelial layer (left panel) with little expression in the stromal layer (right panel). Arrows (white) denote the region of the epithelial–stromal interface. Scale bar = 10 μm [313].
Figure 8. Localization of fibronectin (FN) in corneal epithelial–stromal co-cultures (hCE-TJ/hCF) at 3 days post-airlift. High expression of fibronectin in the epithelial layer (left panel) with little expression in the stromal layer (right panel). Arrows (white) denote the region of the epithelial–stromal interface. Scale bar = 10 μm [313].
Ijms 24 06363 g008
Figure 9. Thrombospondin-1 (TSP-1) expression in corneal epithelial–stromal co-cultures (hCE-TJ/hCF). (A) 3D-reconstruction of the corneal co-culture shows localization of thrombospondin-1 primarily at the epithelial–stromal interface. (B) Individual slices of the epithelial and stromal layers showed high expression within epithelial cells (left panel) and a fibrous appearance of thrombospondin-1 at the epithelial–stromal interface (right panel). Arrows (white) denote the region of the epithelial–stromal interface [313].
Figure 9. Thrombospondin-1 (TSP-1) expression in corneal epithelial–stromal co-cultures (hCE-TJ/hCF). (A) 3D-reconstruction of the corneal co-culture shows localization of thrombospondin-1 primarily at the epithelial–stromal interface. (B) Individual slices of the epithelial and stromal layers showed high expression within epithelial cells (left panel) and a fibrous appearance of thrombospondin-1 at the epithelial–stromal interface (right panel). Arrows (white) denote the region of the epithelial–stromal interface [313].
Ijms 24 06363 g009
Figure 10. Using exosomes for drug delivery. Exosomes are first harvested from the patient. Then, therapeutic agents, including drugs, antibodies, and disease-targeting ligands, can be loaded into exosomes. Finally, the engineered exosomes are re-introduced into the patient.
Figure 10. Using exosomes for drug delivery. Exosomes are first harvested from the patient. Then, therapeutic agents, including drugs, antibodies, and disease-targeting ligands, can be loaded into exosomes. Finally, the engineered exosomes are re-introduced into the patient.
Ijms 24 06363 g010
Table 1. Advantages and disadvantages of exosomes derived from different sources.
Table 1. Advantages and disadvantages of exosomes derived from different sources.
Whole Saliva or Type of Non-SEDisadvantage of Whole Saliva/Non-SE Advantage of SEsShared Disadvantages of SEs and Non-SEs
Whole SalivaHarbors contaminating elements and higher amylase enzyme levelsSE have a lipid bilayer that protects their cargo from contamination and degradation
Proteins from whole saliva are susceptible to degradation when removed from their natural environment
Serum-derived exosomeCollection requires trained personnel and is often more invasive SE collection is easy and non-invasive, improving patient complianceLack of methodologies to quantify contents of exosomes
Coagulation poses challenges in handling these exosomesSaliva does not coagulate
Urinary exosomeApplications limited to kidney and prostate pathologiesWide-ranging applications of SE, including systemic autoimmune disease, neurodegenerative disease, neoplasms, and ocular disease
MSC-derived exosome Rapid clearance from blood after administration in vivo Stable in biological fluids
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

Liu, A.; Hefley, B.; Escandon, P.; Nicholas, S.E.; Karamichos, D. Salivary Exosomes in Health and Disease: Future Prospects in the Eye. Int. J. Mol. Sci. 2023, 24, 6363. https://doi.org/10.3390/ijms24076363

AMA Style

Liu A, Hefley B, Escandon P, Nicholas SE, Karamichos D. Salivary Exosomes in Health and Disease: Future Prospects in the Eye. International Journal of Molecular Sciences. 2023; 24(7):6363. https://doi.org/10.3390/ijms24076363

Chicago/Turabian Style

Liu, Angela, Brenna Hefley, Paulina Escandon, Sarah E. Nicholas, and Dimitrios Karamichos. 2023. "Salivary Exosomes in Health and Disease: Future Prospects in the Eye" International Journal of Molecular Sciences 24, no. 7: 6363. https://doi.org/10.3390/ijms24076363

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