Next Article in Journal
Genomic SELEX Reveals Pervasive Role of the Flagella Master Regulator FlhDC in Carbon Metabolism
Next Article in Special Issue
Mir-302a/TWF1 Axis Impairs the Myogenic Differentiation of Progenitor Cells through F-Actin-Mediated YAP1 Activation
Previous Article in Journal
Genome-Wide Identification of the ERF Transcription Factor Family for Structure Analysis, Expression Pattern, and Response to Drought Stress in Populus alba × Populus glandulosa
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tear Film MicroRNAs as Potential Biomarkers: A Review

1
Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
2
Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
3
Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(4), 3694; https://doi.org/10.3390/ijms24043694
Submission received: 20 December 2022 / Revised: 6 February 2023 / Accepted: 7 February 2023 / Published: 12 February 2023

Abstract

:
MicroRNAs are non-coding RNAs that serve as regulatory molecules in a variety of pathways such as inflammation, metabolism, homeostasis, cell machinery, and development. With the progression of sequencing methods and modern bioinformatics tools, novel roles of microRNAs in regulatory mechanisms and pathophysiological states continue to expand. Advances in detection methods have further enabled larger adoption of studies utilizing minimal sample volumes, allowing the analysis of microRNAs in low-volume biofluids, such as the aqueous humor and tear fluid. The reported abundance of extracellular microRNAs in these biofluids has prompted studies to explore their biomarker potential. This review compiles the current literature reporting microRNAs in human tear fluid and their association with ocular diseases including dry eye disease, Sjögren’s syndrome, keratitis, vernal keratoconjunctivitis, glaucoma, diabetic macular edema, and diabetic retinopathy, as well as non-ocular diseases, including Alzheimer’s and breast cancer. We also summarize the known roles of these microRNAs and shed light on the future progression of this field.

1. Background

MicroRNAs (miRNAs) are a family of short (18–22 nucleotides), non-coding RNAs that serve as significant regulators of overall gene expression [1]. miRNAs released into the extracellular space are capable of providing dynamic insight into cell-to-cell communication, as well as physiological and pathological processes [2,3]. These extracellular miRNAs are detected in biofluids, and their presence in these fluids can be used for diagnostic purposes, giving them significant biomarker potential [4,5,6,7]. miRNAs in circulation have also shown great stability and resistance to degradation over long-term storage and multiple freeze-thaw cycles, which makes them ideal choices for clinical use [8,9]. Furthermore, the nature of miRNAs, with their simpler complexity and straightforward detection/amplification methods, make them a cost-effective choice for the development of established biomarkers compared to their proteomic counterparts [10,11].
Easily accessible biological fluids, such as tears, present exciting potential for future biomarker research. This biofluid is abundant in various molecules and has been shown to contain significantly more unique miRNAs than aqueous humor samples [12]. Advances in miRNA quantitation, paired with the simple, non-invasive collection methods of tears [13], entice mass participant collection [14] and physician engagement [15], which is essential for biomarker validation. Moreover, the importance of studying the relationship between tear composition and ocular health has become more apparent in recent years, and a multitude of studies have reported variations in tear miRNA content across different pathologies [12,16,17,18,19,20,21,22]. The cohesive relationship between miRNAs, the tear film, and health makes tear miRNAs pertinent options as noninvasive and low-cost diagnostic markers for numerous pathologies [13,23,24,25]. Several studies have identified tear miRNAs as potential biomarkers for ocular diseases including diabetic retinopathy [18,26,27], Sjögren’s syndrome [19], glaucoma [20,28], dry eye disease [21,29,30], diabetic macular edema [12], herpes epithelial keratitis [22], and vernal keratoconjunctivitis [31]. Furthermore, tear miRNA alterations have been associated with some non-ocular diseases, such as Alzheimer’s [16] and metastatic breast cancer [32].
This review summarizes the current understanding of miRNAs detected in human tears and their association with both ocular and non-ocular diseases. To date, ~300 unique miRNAs have been reported in tear fluid, of which several are novel and lack in-depth knowledge of their regulatory roles [10,12,16,33]. A comprehensive list of these publications, along with the tear collection and miRNA detection methods used, are presented in Table 1. The general workflow for tear miRNA analysis used in these studies is shown in Figure 1. It is noteworthy that a limited number of studies have examined miRNAs in tears, and therefore more studies with larger sample sets are needed for biomarker validation.

2. Dry Eye Disease

Dry Eye Disease (DED) is a multifactorial disease affecting both the tear film and ocular surface that results in tear film instability and may lead to ocular surface damage [34,35]. The diagnosis of DED could be time-consuming and delayed due to the variability in disease severity, required detailed examinations, and evaluation of history records [36]. Because the tear film is directly altered in this disease, insight into how DED affects its composition can yield novel targets for both diagnosis and treatment.
Searching through the literature, only two studies on profiling tear miRNAs in DED patients have been reported. In one study, nine tear miRNAs (miR-127-5p, miR-1273h-3p, miR-1288-5p, miR-130b-5p, miR-139-3p, miR-1910-5p, miR-203b-5p, miR-22-5p, and miR-4632-3p) were linked to inflammation and were upregulated in the tears of DED patients [29]. The other study reported that four miRNAs (450b-5p, miR-1283, miR-5700, and miR-3671) were significantly upregulated in tears from dry eye patients compared to healthy controls, while 28 tear miRNAs (miR-4673, miR-890, miR-576-5p, miR-337-3p, miR-607, miR-1827, miR-485-5p, miR-6884-5p, miR-335-3p, miR-647, miR-4433b-5p, miR-570-3p, miR-1276, miR-2054, miR-802, miR-539-5p, miR-198, miR-3646, miR-4427, miR-4705, miR-606, miR-20b-3p, miR-4475, miR-4478, miR-4797-5p, miR-142-5p, miR-153-5p, and miR-5583-5p) were significantly downregulated [21]. It is worth noting that there is no overlap in the significantly altered tear miRNAs in DED reported from these two studies. This discrepancy may be attributed to the different pipelines utilized (microarray hybridization vs. RNA-Seq), different collection methodologies (saline wash vs. capillary tubes), and, most importantly, the differences in sample sizes (n = 5 per group: Pucker et al. [29] vs. n = 138 per group: Wang et al. [21]). All of these parameters can contribute to the discrepancies across the literature and examining the impact of each may offer more insight into how to improve reproducibility in future studies exploring this topic. Also, due to high interpersonal variability in tear miRNA levels, studies with a larger sample size are necessary to generate reproducible findings.
Most studies related to these miRNAs (i.e., miR-1288-5p, miR-130b-5p, miR-139-3p, miR-22-5p, miR-203b-5p, and miR-450b-5p) are limited to their involvement in cancer [37,38,39,40,41,42]. One study detected elevated expression of miR-1910-5p in human retinal pigment epithelium cells (APRE-19) in the presence of hydrogen peroxide-induced oxidative stress [43]. Environmental factors, such as exposure to ultraviolet radiation and pollutants, are known to increase oxidative stress and ocular surface inflammation in DED [44]. miR-127, which was elevated in tears from DED patients, has also been shown to promote pro-inflammatory M1 macrophage development in pulmonary tissue of murine models simulating lung inflammation [45]. Higher expression of miR-203 in monkey tears versus sera highlights a potential involvement of miR-203 in ocular homeostasis [46]. Decreased viability of human corneal epithelial (HCE-T) cells has been associated with elevated miR-203 expression compared to basal levels and may be a potential source for its detection in tears [46]. In addition, increased expression of miR-203 in human synovial fibroblasts has been shown to boost the secretion of MMP-1 and IL-6, which are commonly involved in chronic inflammation [47,48,49]. In another study, miR-450b-5p was found to negatively regulate Pax6 in the corneal lineage specification [50]. Pax6 is required for normal eye development [51,52]. However, the precise molecular mechanisms of these miRNAs in DED are still unknown and further studies are required to uncover their roles in an ocular environment.

3. Sjögren’s Syndrome

Sjögren’s syndrome (SS) is a chronic autoimmune disorder characterized by lymphocytic infiltration of exocrine glands, especially the salivary and lacrimal glands [53]. Currently, diagnosis typically relies on a combination of patient-reported symptoms of dry mouth/eyes, measurements of ocular and oral dryness, as well as antinuclear antibody blood tests. Many of these signs and symptoms overlap with those of other inflammatory diseases, making diagnosis difficult; often, this delays treatment. The discovery of reliable tear biomarkers of SS can lead to a faster diagnosis and help prevent mismanagement.
In a recent study, four miRNAs were significantly upregulated (miR-16-5p, miR-34a-5p, miR-142-3p, and miR-223-3p) and ten were significantly downregulated (miR-30b-5p, miR-30c-5p, miR-30d-5p, miR-92a-3p, miR-134-5p, miR-137, miR-302d-5p, miR-365b-3p, miR-374c-5p, miR-487b-3p) in patients with SS compared to healthy controls [19]. However, in this study, the expressions of 43 pre-selected miRNAs were measured using a custom PCR array and thus may not encapsulate all the tear miRNAs significantly altered in SS.
Many of the reported roles of these miRNAs involve the regulation of inflammatory pathways. miR-16 and miR-223 have been reported to be elevated in minor salivary gland tissue and peripheral blood mononuclear cells of SS patients [54]. miR-16 has also been linked to decreased salivary flow rates [55]. Furthermore, the miR-16, miR-142, and miR-223 families are crucial during hematopoietic lineage differentiation and are known to participate in the development and differentiation of immune cells [19,56]. Upregulation of miR-223-3p leads to increased detection of pro-inflammatory cytokines TNF-α and IL-1β in the corneas of mouse models of fungal keratitis [57]. Cortes-Troncoso et al. found that miR-142-3p, which was expressed in the salivary glands of SS patients but not healthy controls, is transported from activated T-cells into glandular cells where it is capable of restricting cAMP production and altering intracellular Ca2+ signaling [58]. Both of these processes are critical in fluid and enzyme secretion from exocrine glands [58]. Therefore, the attenuation of elevated miR-223, miR-16, and miR-142 levels may alleviate the heightened immune response in SS patients and relieve symptoms. However, further studies are required to determine their regulatory involvement.
mir-34a has been associated with regulatory networks in T cell activation [59]. The rapid induction of miR-34a expression in T-cell receptors decreased the killing capacity of primary CD4+ and CD8+ T cells and ultimately shut down the NF-κB signaling process through a proposed feedback loop [60]. Increased activity of CD4+ and CD8+ T cells are common in peripheral circulation and target tissues of SS patients [61,62], and dysfunction of this miRNA pathway may contribute to the damaged glandular epithelial cells found with severe cytotoxic CD8+ T cell infiltration of the exocrine glands and account for the increased miR-34a expression detected in the tears of SS patients.
Also, miR-92 downregulation is associated with the overexpression of innate defense genes in human corneal epithelial cells in response to excessive inflammation [63]. Furthermore, miR-92a expression in minor salivary gland tissue samples from SS patients and healthy volunteers was inversely correlated with SS severity [64]. However, murine models with elevated miR-17-92 cluster expression in their lymphocytes developed lymphoproliferative disease and autoimmunity with enhanced proliferation of both T and B cells [65].
The miR-30 family, which is a known negative regulator of B-cell activating factor (BAFF) [66], was downregulated in the tears of SS patients. BAFF is involved in B-cell maturation, proliferation, and survival, and murine studies have demonstrated that excessive BAFF expression leads to autoimmune-like manifestations [67]. Also, upregulation of BAFF has been observed in B lymphocytes infiltrating the salivary glands of patients with primary SS [66].

4. Herpes Epithelial Keratitis

Herpes epithelial keratitis (HEK) is caused by a herpes simplex virus (HSV) infection of the corneal epithelium, which eventually progresses to form dendritic lesions and geographic ulcers [68]. This can result in a loss of corneal sensation, photophobia, and visual impairment due to corneal scarring [69].
A recent study analyzed the tear miRNA profile of patients with HEK [22] and found that a total of 23 miRNAs were significantly increased in the tears of patients with HEK compared to controls: miR-15b-5p, miR-16-5p, miR-20b-5p, miR-21-5p, miR-23b-3p, miR-25-3p, miR-29a-3p, miR-30a-3p, miR-30d-5p, miR-92a-3p, miR-124-3p, miR-127-3p, miR-132-3p, miR-142-3p, miR-145-5p, miR-146a-5p, miR-146b-5p, miR-155-5p, miR-182-5p, miR-183-5p, miR-221-3p, miR-223-3p, and miR-338-5p. Furthermore, the expression of miR-29a-3p was found to be significantly increased in the dendritic ulcer group compared to the geographic ulcer groups of the HEK patients [22]. This was only a preliminary study with a small sample size that measured the expression of 43 pre-selected miRNAs in tears from eight patients with HEK and seven age-matched controls. Therefore, the miRNA list presented above may not include all the tear miRNAs changing in relation to HEK.
Increased expression of tear miR-223-3p in HEK patients has also been reported in the corneal stromal cells of fungal keratitis murine models [57], associating it with keratitis pathophysiology. Studies have shown upregulation of miR-155 and miR-132 in association with HSV keratitis murine models, specifically in angiogenesis of the cornea [70,71]. Increased vascular endothelial growth factor (VEGF) and subsequent ocular neovascularization in ocular tissues have been associated with HSV infection [72,73]. Interestingly, VEGF has been shown to upregulate the expression of miR-132 in mice corneas after HSV-1 infection, and in vivo silencing of miR-132 in HSV-infected mice correlated with a reduction in corneal neovascularization [74]. The inactivation of miR-183/92/182 using knockout mice significantly decreased corneal inflammation and the severity of microbial keratitis [75]. Another study reported the involvement of miR-183/92/182 expression on pathways such as IL-17 and IL-10 modulation, TLR4 signaling, and activity of corneal macrophages in miR-183/92/182 knockout mice infected with P. aeruginosa that supports the notion of miR-183/92/182 in innate immunity and may contribute to the inflammatory damage experienced in HEK cases [76,77].
Significant upregulation of miR-142-3p has been reported in the tears of SS patients as well as in post-transplant corneas of fungal keratitis patients [78]. This elevated expression in corneal tissue may offer insight into a possible source for this miRNA in tears. miR-142-3p has been shown to inhibit the pro-inflammatory TLR-signaling pathway [79] and may be elevated in response to the corneal inflammation caused by HEK. Also, miR-16 has been shown to be involved in Bcl-2 suppression, and the upregulation in tears of HEK patients may be in response to viral-induced apoptosis [80]. The reported increase in expression of miR-29a-3p in dendritic ulcer groups compared to geographical ulcer groups may signal decreased viral replication, as the miR-29a expression has been shown to interfere with viral Nef protein expression and HIV-1 expression in human HEK293T cells [81]. Dysfunction of this pathway may lead to increased viral turnover and would support the notion of continued viral replication leading to the progression of dendritic ulceration into geographical ulceration [82]. Monitoring of this miRNA may provide insight into the development of HEK and exemplifies the importance of proper patient classification in describing disease-staged miRNA fluctuations.

5. Vernal Keratoconjunctivitis

Vernal Keratoconjunctivitis (VKC) is a chronic, inflammatory condition of the conjunctiva with a pathogenesis that remains unclear [83]. While the disease is self-limited, its implications can cause corneal scarring and severe visual impairment [84]. A total of 51 tear miRNAs were differentially expressed in VKC patients. The expressions of miR-1229-5p, miR-6821-5p, miR-6800-5p, miR-4466, miR-3665, miR-4530, miR-7110-5p, miR-1207-5p, miR-6875-5p, miR-762, miR-4741, miR-6740-5p, and miR-4298 were significantly upregulated and the expressions of miR-7975, miR-7977, and miR-1260a were significantly downregulated in the VKC patients compared to healthy controls [31]. However, the small sample size (n = 4 per group) may limit the reproducibility of these findings.
miR-4530 has been associated with the suppression of cell proliferation and the regulation of inflammatory processes [85,86]. The detected upregulation of this miRNA in VKC patient tears may be in response to pro-inflammatory species and stressors present during the disease. Upregulated miR-762 has been shown to negatively regulate host defense genes in human corneal epithelial cells exposed to external antigens [63]. This may contribute to a dysfunctional corneal epithelial layer that allows increased allergen uptake, which can lead to the increased activation of inflammatory responses [87,88]. This study also found upregulated expression of corneal epithelial miR-1207 in response to bacterial antigens, suggesting miR-1207’s involvement in host defense mechanisms. miR-4466 was found to be overexpressed in human corneal stromal cell exosomes of patients with keratoconus compared to healthy controls [89]. This upregulation was also detected in the tears from VKC patients and may indicate miR-4466’s potential involvement in corneal surface integrity.

6. Primary Open-Angle Glaucoma

Primary open-angle glaucoma (POAG) is associated with diminished aqueous outflow and cupping of the optic nerve head, which can lead to neuro-retinal degeneration and eventual blindness [90]. When comparing tear miRNA levels in POAG subjects with controls, the expression of miR-26b-5p, miR-27a-3p, miR-152-3p, miR-30e-5p, miR-125b-2-5p, and miR-224-5p was increased in POAG patients, while the expression of miR-151a-3p and miR-1307-3p was decreased [20]. Another study detected decreased levels of miR-146b, miR-126, and miR-16 in the tears of POAG patients compared to healthy controls [28]. It is important to note the variability in miRNAs reported between these studies. Tamkovich et al. [28] pre-selected miR-146b, miR-16, and miR-126 and performed targeted RT-PCR analysis, while Raga-Cervera et al. [20] used RNA-Seq, which would explain the greater number of unique miRNAs reported. Eight miRNAs were significantly altered using RNA-Seq, but the findings of these studies do not overlap. Therefore, other factors such as control group parameters and subject variability may have contributed to this difference.
Many of these miRNAs have been previously associated with glaucoma pathogenesis. miR-125b expression was reported significantly lower in aqueous humor samples of POAG patients [91]. miR-27a, which is believed to have neuroprotective capabilities via the inhibition of proapoptotic Bcl-2 protein synthesis during neuronal cell death [92], was found to be significantly upregulated in glaucomatous rodent retinas [93].
Ren et al. have proposed that miR-27a plays a regulatory role in oxidative stress-induced apoptosis of retinal pigment epithelium and retinal degeneration [94]. The miR-27a-induced inhibition of FOXO1-related autophagic functions modulates reactive oxygen species (ROS) accumulation in the retinal pigment epithelium and eventually leads to retinal breakdown [94]. ROS accumulation is a key component in the pathogenesis of POAG [95]. miR-125b, a microglia-enriched miRNA shown to cause constitutive NF-κB activation via suppressing zinc-finger protein A20 expression [96], may be relevant to the retinal neurodegeneration experienced in glaucoma patients from excited microglia [97]. Conversely, members of the miR-146 family are known negative regulators of the inflammatory pathway NF-κB through the suppression of thrombin-induced GPCR-mediated NF-κB activation in human retinal endothelial cells and inhibition of diabetes-induced NF-κB activation and retinal microvascular in diabetic rat models [98,99]. Furthermore, reports show that miR-146 is involved in the suppressive roles of COX-2 and IL-1β expression in human endothelial cells, epithelial cells, fibroblasts, and monocytes [100,101,102]. IL-1β expression has been shown in human optic nerves and reported significantly elevated in AH samples of POAG patients compared to controls [103]. With significantly increased oxidative stress reported in the aqueous humor of POAG patients, downregulation of tear miR-146b may signal its dysregulation to be a component of POAG pathophysiology [104].
On the contrary, miR-16 has been associated with the inhibition of cell proliferation and the induction of apoptosis due to its involvement with the suppression of the anti-apoptotic Bcl-2 gene [80]. With an increased presence of cell apoptosis in the trabecular meshwork of POAG patients [105], the down-regulation of miR-16 may indicate an upstream regulatory attempt at preventing cell apoptosis during glaucoma [28]. While reduced expression of miR-126 was reported in the tears of POAG patients [28], elevated miR-126 expression was reported in murine models of glaucoma [106]. The study also found that miR-126 facilitates apoptosis of retinal ganglion cells by promoting the VEGF-Notch pathway in murine models of glaucoma [106]. However, another study reported that pretreating monkey chorioretinal vessel endothelial cells with miR-126-mimics sharply decreased VEGF expression levels [107].

7. Diabetic Macular Edema

Diabetic macular edema (DME) leads to the breakdown of the blood-retinal barrier, resulting in macular thickening and intra-retinal leakage of fluids, proteins, and lipids [108]. In a recent study, the tear expression of twelve tear miRNAs was significantly correlated with either a good or a poor response to anti-VEGF treatment in DME patients. The expression of miR-214-3p, miR-320d, and miR-874-3p was positively correlated with a decrease in retinal central subfield thickness (CST) in response to anti-VEGF treatment while miR-98-5p, miR-196b-5p, and miR-454-3p correlated with increases in retinal CST following treatment [12]. Although limited by small group sizes (n = 6, n = 7, and n = 11), this study detected a greater number of unique miRNAs (331) in tears compared to serum (327) and aqueous humor samples (231).
miR-214 and miR-874 have been shown to suppress oxidative stress in diabetes and alleviate diabetes-induced nerve injury and inflammation [109,110,111]. The miR-320 family has been found to negatively regulate VEGF expression during high glucose exposure in human vein endothelial cells [112]. The upregulation of miR-320 in tear samples of DME patients may be in response to elevated glucose levels as studies have shown a correlation between the glucose content of plasma and tear fluid [113].

8. Diabetic Retinopathy

Diabetic retinopathy (DR) is a microvascular disease that can lead to irreversible vision impairment. As its prevalence continues to grow, there is a clear need for improved early detection and diagnosis. Tear miRNA composition has been shown to be altered in DR patients. Sun et al. found that miR-23a expression was decreased in the tears and serum of patients with type 2 diabetes or pre-diabetes compared to controls [18]. This is intriguing as miR-23a has been linked to a potential inhibitory role on VEGF, an important factor in the development of DR and blood vessel growth [114,115]. Hu et al. found elevated expression of miR-9-5p, miR-143-3p, miR-411-5p, miR-218-5p, miR-139-5p, miR-10b-5p, miR-129-5p, miR-214-3p, miR-381-3p, miR-130b-4p, miR-382-5p, miR-145-5p, miR-379-5p, miR-127-3p, miR-196a-5p, miR-206, miR-1-3p, miR-199a-5p, miR-642a-3p, and miR-642b-5p and decreased expression of miR-361-3p, miR-29c-5p, miR-532-5p, miR-3615, miR-429, miR-224-5p, miR-146a-5p, miR-149-5p, miR-339-3p, miR-203a-3p, miR-24-1-5p, miR-203b-5p, miR-31-5p, miR-147b-3p, miR-205-5p, miR-186-5p, miR-7706, miR-191-5p, miR-135b-5p, and miR-96-5p in tears of DR patients compared to healthy controls. It was also reported that the expression of miR-218-5p continuously increased from healthy controls to diabetic patients with and without DR and may be a potential biomarker for disease progression [27]. Sun et al. pre-selected miRNA-23a for RT-PCR analysis, while Hu et al. used RNA-Seq. However, miR-23a was not found to be decreased in the RNA-Seq study. Several factors may have led to this discrepancy. It is also important to mention that, while the mean ages of DR patients were similar between studies (53.6 vs. 54.6 years old), the mean age of the healthy controls differs (30.3 vs. 55.2 years old), which could impact the findings. Additionally, proteomic studies have found that the different tear sample collection techniques used in these two studies (Schirmer strips vs. capillary tubes) impact the identified tear composition [116]. It is possible that different collection methods have similar effects on identified miRNA content.
miR-145-5p expression has been shown to be elevated in high glucose-induced retinal ganglion cells (RGC) and linked to increased pro-inflammatory cytokines, IL-6, and apoptosis [117]. Another study found silencing of miR-145-5p inhibited apoptosis of RGCs via TRIM2-mediated activation of the PI3K/AKT signaling pathway in glaucoma rat models [118]. However, other literature has reported decreased expression of miR-145 in high glucose-treated retinal endothelial cells, making it apparent that more studies are needed to uncover miR-145’s role in various cell types [119]. Both miR-218-5p and miR-9-5p have been linked to insulin pathways [120,121]. miR-9 has also been reported to be significantly elevated in the serum of diabetic patients compared to controls and may serve as a potential biomarker for diabetes susceptibility [122]. Interestingly, miR-146 expression was also reportedly downregulated in the tears of glaucoma patients, but upregulated in the tears of HEK patients. The involvement of miR-146 in inflammatory pathways may signal dysfunctional inflammatory mechanisms that propagate DR or POAG symptoms as opposed to the herpes-induced complications found in HEK. Intraocular injection of pre-miR-31 has been shown to reduce retinal neovascularization and its absence in tears of DR patients may signal dysregulated pathways leading to its development [123]. Elevated miR-96 expression in RGC-5 cells showed significant hindrance to RGC-5 survival rates, and its reported decrease in DR tears may correlate with an attempt to prevent further damage [124].

9. Alzheimer’s Disease

Alzheimer’s disease (AD) is a neurodegenerative disease characterized by the progressive impairment of cognitive function [125]. Alterations in tear composition and flow rate have been associated with AD, and certain miRNAs, such as miR-15, have been linked to the inhibition of tau phosphorylation in mouse neuronal cells [16,126,127]. A recent study compared the tear miRNA profile of AD patients to healthy controls and found overlap of 133 miRNAs common between sample groups, of which 90 displayed altered expression levels. One miRNA, miR-200b-5p, was selected as a potential biomarker as it was exclusively detected in tear samples from AD patients [16]. Interestingly, miR-200b/c has been shown to be upregulated with the reduction of amyloid-β secretion and amyloid-β–induced memory impairment in primary neurons of AD murine models [128]. Another study showed the downregulation of miR-200b in the hippocampi of amyloid-precursor protein transgenic mice compared to controls [129]. The detection of miR-200b-5p in the tears of AD patients provides a potential biomarker and therapeutic target.

10. Breast Cancer

While there have been great improvements in the diagnosis and treatment of metastatic breast cancer over the years, 5-year survival remains low [130,131]. Detection at the earliest stages is paramount to preventing disease progression, and the search for novel diagnostic methods continues to expand. miRNAs have become widely accepted as important components in a variety of pathways involved in regulating breast cancer [132]. However, non-invasive means of collection and quantification are limited. A study reported the expression of both miR-21 and miR-200c to be significantly higher in tear exosomes of patients with metastatic breast cancer compared to healthy controls [32]. It should be noted that the authors pre-selected these miRNAs for tear analysis as upregulation of these miRNAs has also been observed in the plasma of metastatic breast cancer patients compared to subjects with localized breast cancer and healthy controls [133,134,135]. A comprehensive tear miRNA profiling of metastatic breast cancer patients may provide additional miRNA biomarkers.
miR-21 is frequently overexpressed in various human tumors [136], and functional studies have reported its involvement in oncogenic processes such as high proliferation [137], low apoptosis [138], and metastatic potential [139]. Interestingly, another study reported a strong association of elevated serum miR-200c expression with colorectal cancer metastasis [140]. Jurmeister et al. showed elevated miR-200c levels repress the migration and invasion of human breast cancer cells by targeting actin-regulatory proteins [141]. This may correlate to the elevated detection of miR-200c in the tears of metastasis breast cancer patients.

11. Overall Summary

Compared to other biofluids like blood, serum, cerebrospinal fluid, saliva, and synovial fluid, a limited number of studies have been conducted so far on tear film miRNAs. With recent advances in detection methods and bioinformatics tools, studies exploring tear film miRNAs are gaining more traction due to their extracellular stability, tissue specificity, and posttranslational regulatory capabilities. As easily accessible biomarkers, tear miRNAs can provide a necessary window into the homeostatic state of the body and offer both biomarker and therapeutic potential. The present review provided an insight into the current findings of tear film miRNAs and their association with dry eye disease, Sjögren’s syndrome, herpes epithelial keratitis, vernal keratoconjunctivitis, primary open-angle glaucoma, diabetic macular edema, diabetic retinopathy, Alzheimer’s, and metastatic breast cancer. A complete list of the tear miRNAs and their biomarker potential in relation to these diseases is shown in Table 2.

12. Concluding Remarks and Future Directions

Due to the limited amount of research in the field, it is difficult to establish the baseline miRNA profile of human tears. The lack of standardized methods limit reproducibility and hinder the comparison of results between studies. Consequently, further studies using both discovery and targeted workflows are needed to uncover the biomarker potential of tear miRNAs.
Further, the expression and function of these molecules are known to be tissue-specific, which makes it challenging to extrapolate their regulatory mechanisms across a variety of tissues. Interpersonal differences and daily fluctuations of miRNAs also pose issues for controlled studies. The maturation of this field relies on the establishment of a proven means of collection, extraction, and analysis that can reduce the variability between studies and make large-scale collaboration feasible. Extensive examination of patient medical history can facilitate better categorization of samples, which will allow the identification of fluctuations in miRNA levels due to demographic variation and different stages of disease. Promoting follow-up studies with larger cohorts will help overcome the analytical difficulties posed by variability in miRNA expression. In these circumstances, effective sampling and analytical techniques are crucial, and improving data storage and sharing is essential to establishing the framework for tear miRNAs as biomarkers.

Author Contributions

Conceptualization, J.A., G.J., S.S. and A.S.; investigation, J.A., G.J. and A.S.; writing—original draft preparation, J.A., G.J. and S.A.; writing—review and editing, A.S. and S.S.; visualization, S.A., J.A., G.J. and A.S.; supervision, A.S. and S.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Institutes of Health, National Eye Institute (Bethesda, MD, USA) grant# R01 EY029728 awarded to Ashok Sharma, grant# R01 EY026936 awarded to Shruti Sharma, and grant# P30 EY031631 Center Core Grant for Vision Research.

Data Availability Statement

Data sharing not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hammond, S.M. An overview of microRNAs. Adv. Drug Deliv. Rev. 2015, 87, 3–14. [Google Scholar] [CrossRef]
  2. O’Brien, J.; Hayder, H.; Zayed, Y.; Peng, C. Overview of microRNA biogenesis, mechanisms of actions, and circulation. Front. Endocrinol. 2018, 9, 402. [Google Scholar] [CrossRef]
  3. Conti, I.; Varano, G.; Simioni, C.; Laface, I.; Milani, D.; Rimondi, E.; Neri, L.M. miRNAs as influencers of cell–cell communication in tumor microenvironment. Cells 2020, 9, 220. [Google Scholar] [CrossRef] [PubMed]
  4. Lawrie, C.H.; Gal, S.; Dunlop, H.M.; Pushkaran, B.; Liggins, A.P.; Pulford, K.; Banham, A.H.; Pezzella, F.; Boultwood, J.; Wainscoat, J.S. Detection of elevated levels of tumour-associated microRNAs in serum of patients with diffuse large B-cell lymphoma. Br. J. Haematol. 2008, 141, 672–675. [Google Scholar] [CrossRef] [PubMed]
  5. Wang, J.; Chen, J.; Sen, S. MicroRNA as biomarkers and diagnostics. J. Cell. Physiol. 2016, 231, 25–30. [Google Scholar] [CrossRef] [PubMed]
  6. Hrašovec, S.; Glavač, D. MicroRNAs as novel biomarkers in colorectal cancer. Front. Genet. 2012, 3, 180. [Google Scholar] [CrossRef]
  7. Etheridge, A.; Lee, I.; Hood, L.; Galas, D.; Wang, K. Extracellular microRNA: A new source of biomarkers. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2011, 717, 85–90. [Google Scholar] [CrossRef] [PubMed]
  8. Van den Berg, M.; Krauskopf, J.; Ramaekers, J.; Kleinjans, J.; Prickaerts, J.; Briedé, J. Circulating microRNAs as potential biomarkers for psychiatric and neurodegenerative disorders. Prog. Neurobiol. 2020, 185, 101732. [Google Scholar] [CrossRef]
  9. Matias-Garcia, P.R.; Wilson, R.; Mussack, V.; Reischl, E.; Waldenberger, M.; Gieger, C.; Anton, G.; Peters, A.; Kuehn-Steven, A. Impact of long-term storage and freeze-thawing on eight circulating microRNAs in plasma samples. PLoS ONE 2020, 15, e0227648. [Google Scholar] [CrossRef] [PubMed]
  10. Weber, J.A.; Baxter, D.H.; Zhang, S.; Huang, D.Y.; How Huang, K.; Jen Lee, M.; Galas, D.J.; Wang, K. The microRNA spectrum in 12 body fluids. Clin. Chem. 2010, 56, 1733–1741. [Google Scholar] [CrossRef] [Green Version]
  11. Xi, X.; Li, T.; Huang, Y.; Sun, J.; Zhu, Y.; Yang, Y.; Lu, Z.J. RNA biomarkers: Frontier of precision medicine for cancer. Non-Coding RNA 2017, 3, 9. [Google Scholar] [CrossRef] [PubMed]
  12. Chan, H.W.; Yang, B.; Wong, W.; Blakeley, P.; Seah, I.; Tan, Q.S.W.; Wang, H.; Bhargava, M.; Lin, H.A.; Chai, C.H. A pilot study on microRNA profile in tear fluid to predict response to anti-VEGF treatments for diabetic macular edema. J. Clin. Med. 2020, 9, 2920. [Google Scholar] [CrossRef] [PubMed]
  13. Bachhuber, F.; Huss, A.; Senel, M.; Tumani, H. Diagnostic biomarkers in tear fluid: From sampling to preanalytical processing. Sci. Rep. 2021, 11, 10064. [Google Scholar] [CrossRef] [PubMed]
  14. Quah, J.H.M.; Tong, L.; Barbier, S. Patient acceptability of tear collection in the primary healthcare setting. Optom. Vis. Sci. 2014, 91, 452. [Google Scholar] [CrossRef]
  15. Posa, A.; Bräuer, L.; Schicht, M.; Garreis, F.; Beileke, S.; Paulsen, F. Schirmer strip vs. capillary tube method: Non-invasive methods of obtaining proteins from tear fluid. Ann. Anat.-Anat. Anz. 2013, 195, 137–142. [Google Scholar] [CrossRef]
  16. Kenny, A.; Jiménez-Mateos, E.M.; Zea-Sevilla, M.A.; Rábano, A.; Gili-Manzanaro, P.; Prehn, J.H.; Henshall, D.C.; Ávila, J.; Engel, T.; Hernández, F. Proteins and microRNAs are differentially expressed in tear fluid from patients with Alzheimer’s disease. Sci. Rep. 2019, 9, 15437. [Google Scholar] [CrossRef] [PubMed]
  17. López-López, M.; Regueiro, U.; Bravo, S.B.; del Pilar Chantada-Vázquez, M.; Varela-Fernández, R.; Ávila-Gómez, P.; Hervella, P.; Lema, I. Tear proteomics in keratoconus: A quantitative SWATH-MS analysis. Investig. Ophthalmol. Vis. Sci. 2021, 62, 30. [Google Scholar] [CrossRef]
  18. Sun, L.; Liu, X.; Zuo, Z. Regulatory role of miRNA-23a in diabetic retinopathy. Exp. Ther. Med. 2021, 22, 1477. [Google Scholar] [CrossRef]
  19. Kim, Y.J.; Yeon, Y.; Lee, W.J.; Shin, Y.U.; Cho, H.; Sung, Y.-K.; Lim, H.W.; Kang, M.H. Comparison of MicroRNA expression in tears of normal subjects and Sjögren syndrome patients. Investig. Ophthalmol. Vis. Sci. 2019, 60, 4889–4895. [Google Scholar] [CrossRef] [PubMed]
  20. Raga-Cervera, J.; Bolarin, J.M.; Millan, J.M.; Garcia-Medina, J.J.; Pedrola, L.; Abellán-Abenza, J.; Valero-Vello, M.; Sanz-González, S.M.; O’Connor, J.E.; Galarreta-Mira, D. miRNAs and genes involved in the interplay between ocular hypertension and primary open-angle glaucoma. Oxidative stress, inflammation, and apoptosis networks. J. Clin. Med. 2021, 10, 2227. [Google Scholar] [CrossRef]
  21. Wang, Q.; Xie, X.; Li, H.; Hao, S. Discovery of microRNA expression profiles involved in regulating TGF-β 2 expression in the tears of dry eye patients. Ann. Clin. Biochem. 2020, 57, 420–428. [Google Scholar] [CrossRef]
  22. Kim, Y.J.; Yeon, Y.; Lee, W.J.; Shin, Y.U.; Cho, H.; Lim, H.W.; Kang, M.H. Analysis of MicroRNA Expression in Tears of Patients with Herpes Epithelial Keratitis: A Preliminary Study. Investig. Ophthalmol. Vis. Sci. 2022, 63, 21. [Google Scholar] [CrossRef] [PubMed]
  23. Dartt, D.; Willcox, M. Complexity of the tear film: Importance in homeostasis and dysfunction during disease. Exp. Eye Res. 2013, 117, 1. [Google Scholar] [CrossRef]
  24. Hagan, S.; Martin, E.; Enríquez-de-Salamanca, A.J.E.J. Tear fluid biomarkers in ocular and systemic disease: Potential use for predictive, preventive and personalised medicine. EPMA J. 2016, 7, 15. [Google Scholar] [CrossRef]
  25. Kalmodia, S.; Son, K.-N.; Cao, D.; Lee, B.-S.; Surenkhuu, B.; Shah, D.; Ali, M.; Balasubramaniam, A.; Jain, S.; Aakalu, V.K. Presence of histatin-1 in human tears and association with aqueous deficient dry eye diagnosis: A preliminary study. Sci. Rep. 2019, 9, 10304. [Google Scholar] [CrossRef] [PubMed]
  26. Pinazo-Durán, M.; Zanón-Moreno, V.; Lleó-Perez, A.; García-Medina, J.; Galbis-Estrada, C.; Roig-Revert, M.; Marco-Ramírez, C.; López-Gálvez, M.; Dolz-Marco, R.; Duarte, L. Genetic systems for a new approach to risk of progression of diabetic retinopathy. Arch. De La Soc. Española De Oftalmol. Engl. Ed. 2016, 91, 209–216. [Google Scholar] [CrossRef] [PubMed]
  27. Hu, L.; Zhang, T.; Ma, H.; Pan, Y.; Wang, S.; Liu, X.; Dai, X.; Zheng, Y.; Lee, L.P.; Liu, F. Discovering the Secret of Diseases by Incorporated Tear Exosomes Analysis via Rapid-Isolation System: iTEARS. ACS Nano 2022, 16, 11720–11732. [Google Scholar] [CrossRef] [PubMed]
  28. Tamkovich, S.; Grigor’eva, A.; Eremina, A.; Tupikin, A.; Kabilov, M.; Chernykh, V.; Vlassov, V.; Ryabchikova, E. What information can be obtained from the tears of a patient with primary open angle glaucoma? Clin. Chim. Acta 2019, 495, 529–537. [Google Scholar] [CrossRef] [PubMed]
  29. Pucker, A.D.; Ngo, W.; Postnikoff, C.K.; Fortinberry, H.; Nichols, J.J. Tear Film miRNAs and Their Association With Human Dry Eye Disease. Curr. Eye Res. 2022, 47, 1479–1487. [Google Scholar] [CrossRef] [PubMed]
  30. Ren, Y.; Feng, J.; Lin, Y.; Reinach, P.S.; Liu, Y.; Xia, X.; Ma, X.; Chen, W.; Zheng, Q. MiR-223 inhibits hyperosmolarity-induced inflammation through downregulating NLRP3 activation in human corneal epithelial cells and dry eye patients. Exp. Eye Res. 2022, 220, 109096. [Google Scholar] [CrossRef] [PubMed]
  31. Syed, N.H.; Shahidan, W.N.S.; Shatriah, I.; Zunaina, E. MicroRNA Profiling of the Tears of Children with Vernal Keratoconjunctivitis. Front. Genet. 2022, 13, 847168. [Google Scholar] [CrossRef]
  32. Inubushi, S.; Kawaguchi, H.; Mizumoto, S.; Kunihisa, T.; Baba, M.; Kitayama, Y.; Takeuchi, T.; Hoffman, R.M.; Sasaki, R. Oncogenic miRNAs identified in tear exosomes from metastatic breast cancer patients. Anticancer. Res. 2020, 40, 3091–3096. [Google Scholar] [CrossRef] [PubMed]
  33. Ravishankar, P.; Daily, A. Tears as the Next Diagnostic Biofluid: A Comparative Study between Ocular Fluid and Blood. Appl. Sci. 2022, 12, 2884. [Google Scholar] [CrossRef]
  34. Messmer, E.M. The pathophysiology, diagnosis, and treatment of dry eye disease. Dtsch. Ärzteblatt Int. 2015, 112, 71. [Google Scholar] [CrossRef]
  35. Craig, J.P.; Nichols, K.K.; Akpek, E.K.; Caffery, B.; Dua, H.S.; Joo, C.-K.; Liu, Z.; Nelson, J.D.; Nichols, J.J.; Tsubota, K. TFOS DEWS II definition and classification report. Ocul. Surf. 2017, 15, 276–283. [Google Scholar] [CrossRef] [PubMed]
  36. Wolffsohn, J.S.; Arita, R.; Chalmers, R.; Djalilian, A.; Dogru, M.; Dumbleton, K.; Gupta, P.K.; Karpecki, P.; Lazreg, S.; Pult, H. TFOS DEWS II diagnostic methodology report. Ocul. Surf. 2017, 15, 539–574. [Google Scholar] [CrossRef]
  37. Gopalan, V.; Pillai, S.; Ebrahimi, F.; Salajegheh, A.; Lam, T.C.; Le, T.K.; Langsford, N.; Ho, Y.H.; Smith, R.A.; Lam, A.K.Y. Regulation of microRNA-1288 in colorectal cancer: Altered expression and its clinicopathological significance. Mol. Carcinog. 2014, 53, E36–E44. [Google Scholar] [CrossRef] [PubMed]
  38. Zhao, G.; Zhang, J.-G.; Shi, Y.; Qin, Q.; Liu, Y.; Wang, B.; Tian, K.; Deng, S.-C.; Li, X.; Zhu, S. MiR-130b is a prognostic marker and inhibits cell proliferation and invasion in pancreatic cancer through targeting STAT3. PLoS ONE 2013, 8, e73803. [Google Scholar]
  39. Krishnan, K.; Steptoe, A.L.; Martin, H.C.; Pattabiraman, D.R.; Nones, K.; Waddell, N.; Mariasegaram, M.; Simpson, P.T.; Lakhani, S.R.; Vlassov, A. miR-139-5p is a regulator of metastatic pathways in breast cancer. RNA 2013, 19, 1767–1780. [Google Scholar] [CrossRef] [PubMed]
  40. Xu, D.; Takeshita, F.; Hino, Y.; Fukunaga, S.; Kudo, Y.; Tamaki, A.; Matsunaga, J.; Takahashi, R.-U.; Takata, T.; Shimamoto, A. miR-22 represses cancer progression by inducing cellular senescence. J. Cell Biol. 2011, 193, 409–424. [Google Scholar] [CrossRef] [PubMed]
  41. Wang, C.; Wang, X.; Liang, H.; Wang, T.; Yan, X.; Cao, M.; Wang, N.; Zhang, S.; Zen, K.; Zhang, C. miR-203 inhibits cell proliferation and migration of lung cancer cells by targeting PKCα. PLoS ONE 2013, 8, e73985. [Google Scholar] [CrossRef] [PubMed]
  42. Jin, Y.; Jiang, Z.; Guan, X.; Chen, Y.; Tang, Q.; Wang, G.; Wang, X. miR-450b-5p suppresses stemness and the development of chemoresistance by targeting SOX2 in colorectal cancer. DNA Cell Biol. 2016, 35, 249–256. [Google Scholar] [CrossRef] [PubMed]
  43. Ayaz, L.; Dinç, E. Evaluation of microRNA responses in ARPE-19 cells against the oxidative stress. Cutan. Ocul. Toxicol. 2018, 37, 121–126. [Google Scholar] [CrossRef]
  44. Seen, S.; Tong, L. Dry eye disease and oxidative stress. Acta Ophthalmol. 2018, 96, e412–e420. [Google Scholar] [CrossRef] [PubMed]
  45. Ying, H.; Kang, Y.; Zhang, H.; Zhao, D.; Xia, J.; Lu, Z.; Wang, H.; Xu, F.; Shi, L. MiR-127 modulates macrophage polarization and promotes lung inflammation and injury by activating the JNK pathway. J. Immunol. 2015, 194, 1239–1251. [Google Scholar] [CrossRef] [PubMed]
  46. Nakagawa, A.; Nakajima, T.; Azuma, M. Tear miRNA expression analysis reveals miR-203 as a potential regulator of corneal epithelial cells. BMC Ophthalmol. 2021, 21, 377. [Google Scholar] [CrossRef] [PubMed]
  47. Manicone, A.M.; McGuire, J.K. Matrix metalloproteinases as modulators of inflammation. In Seminars in Cell & Developmental Biology; Academic Press: Cambridge, MA, USA, 2008; pp. 34–41. [Google Scholar]
  48. Yao, X.; Huang, J.; Zhong, H.; Shen, N.; Faggioni, R.; Fung, M.; Yao, Y. Targeting interleukin-6 in inflammatory autoimmune diseases and cancers. Pharmacol. Ther. 2014, 141, 125–139. [Google Scholar] [CrossRef]
  49. Hirano, T. IL-6 in inflammation, autoimmunity and cancer. Int. Immunol. 2021, 33, 127–148. [Google Scholar] [CrossRef] [PubMed]
  50. Shalom-Feuerstein, R.; Serror, L.; De La Forest Divonne, S.; Petit, I.; Aberdam, E.; Camargo, L.; Damour, O.; Vigouroux, C.; Solomon, A.; Gaggioli, C.; et al. Pluripotent stem cell model reveals essential roles for miR-450b-5p and miR-184 in embryonic corneal lineage specification. Stem Cells 2012, 30, 898–909. [Google Scholar] [CrossRef]
  51. Ramaesh, T.; Collinson, J.M.; Ramaesh, K.; Kaufman, M.H.; West, J.D.; Dhillon, B. Corneal abnormalities in Pax6+/− small eye mice mimic human aniridia-related keratopathy. Investig. Ophthalmol. Vis. Sci. 2003, 44, 1871–1878. [Google Scholar] [CrossRef]
  52. Li, W.; Chen, Y.-T.; Hayashida, Y.; Blanco, G.; Kheirkah, A.; He, H.; Chen, S.-Y.; Liu, C.-Y.; Tseng, S.C.G. Down-regulation of Pax6 is associated with abnormal differentiation of corneal epithelial cells in severe ocular surface diseases. J. Pathol. 2008, 214, 114–122. [Google Scholar] [CrossRef] [Green Version]
  53. Patel, R.; Shahane, A. The epidemiology of Sjögren’s syndrome. Clin. Epidemiol. 2014, 6, 247. [Google Scholar] [PubMed]
  54. Cha, S.; Mona, M.; Lee, K.E.; Kim, D.H.; Han, K. MicroRNAs in autoimmune Sjögren’s syndrome. Genom. Inform. 2018, 16, e19. [Google Scholar] [CrossRef] [PubMed]
  55. Alevizos, I.; Alexander, S.; Turner, R.J.; Illei, G.G. MicroRNA expression profiles as biomarkers of minor salivary gland inflammation and dysfunction in Sjögren’s syndrome. Arthritis Rheum. 2011, 63, 535–544. [Google Scholar] [CrossRef]
  56. Chen, C.-Z.; Li, L.; Lodish, H.F.; Bartel, D.P. MicroRNAs modulate hematopoietic lineage differentiation. Science 2004, 303, 83–86. [Google Scholar] [CrossRef] [PubMed]
  57. Tang, H.; Lin, Y.; Huang, L.; Hu, J. MiR-223-3p Regulates Autophagy and Inflammation by Targeting ATG16L1 in Fusarium solani–Induced Keratitis. Investig. Ophthalmol. Vis. Sci. 2022, 63, 41. [Google Scholar] [CrossRef] [PubMed]
  58. 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 Sjögren’s syndrome. JCI Insight 2020, 5, e133497. [Google Scholar] [CrossRef]
  59. Taheri, F.; Ebrahimi, S.O.; Shareef, S.; Reiisi, S. Regulatory and immunomodulatory role of miR-34a in T cell immunity. Life Sci. 2020, 262, 118209. [Google Scholar] [CrossRef]
  60. Hart, M.; Walch-Rückheim, B.; Friedmann, K.S.; Rheinheimer, S.; Tänzer, T.; Glombitza, B.; Sester, M.; Lenhof, H.-P.; Hoth, M.; Schwarz, E.C. miR-34a: A new player in the regulation of T cell function by modulation of NF-κB signaling. Cell Death Dis. 2019, 10, 46. [Google Scholar] [CrossRef]
  61. Youinou, P.; Pennec, Y.; Blaschek, M.; Gentric, A.; Jouquan, J.; Lamour, A.; Angelidis, P. Activation of peripheral blood lymphocytes in patients with primary Sjögren’s syndrome. Rheumatol. Int. 1988, 8, 125–130. [Google Scholar] [CrossRef]
  62. Zhou, H.; Yang, J.; Tian, J.; Wang, S. CD8+ T lymphocytes: Crucial players in sjögren’s syndrome. Front. Immunol. 2021, 11, 602823. [Google Scholar] [CrossRef] [PubMed]
  63. Mun, J.; Tam, C.; Chan, G.; Kim, J.H.; Evans, D.; Fleiszig, S. MicroRNA-762 is upregulated in human corneal epithelial cells in response to tear fluid and Pseudomonas aeruginosa antigens and negatively regulates the expression of host defense genes encoding RNase7 and ST2. PLoS ONE 2013, 8, e57850. [Google Scholar] [CrossRef] [PubMed]
  64. Yan, T.; Shen, J.; Chen, J.; Zhao, M.; Guo, H.; Wang, Y. Differential expression of miR-17-92 cluster among varying histological stages of minor salivary gland in patients with primary Sjögren’s syndrome. Clin. Exp. Rheumatol. 2019, 37, 49–54. [Google Scholar]
  65. Xiao, C.; Srinivasan, L.; Calado, D.P.; Patterson, H.C.; Zhang, B.; Wang, J.; Henderson, J.M.; Kutok, J.L.; Rajewsky, K. Lymphoproliferative disease and autoimmunity in mice with increased miR-17-92 expression in lymphocytes. Nat. Immunol. 2008, 9, 405–414. [Google Scholar] [CrossRef] [PubMed]
  66. Wang-Renault, S.-F.; Boudaoud, S.; Nocturne, G.; Roche, E.; Sigrist, N.; Daviaud, C.; Tinggaard, A.B.; Renault, V.; Deleuze, J.-F.; Mariette, X. Deregulation of microRNA expression in purified T and B lymphocytes from patients with primary Sjögren’s syndrome. Ann. Rheum. Dis. 2018, 77, 133–140. [Google Scholar] [CrossRef] [PubMed]
  67. Mackay, F.; Woodcock, S.A.; Lawton, P.; Ambrose, C.; Baetscher, M.; Schneider, P.; Tschopp, J.; Browning, J.L. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J. Exp. Med. 1999, 190, 1697–1710. [Google Scholar] [CrossRef] [PubMed]
  68. Kaye, S.; Choudhary, A. Herpes simplex keratitis. Prog. Retin. Eye Res. 2006, 25, 355–380. [Google Scholar] [CrossRef] [PubMed]
  69. Azher, T.N.; Yin, X.-T.; Tajfirouz, D.; Huang, A.J.; Stuart, P.M. Herpes simplex keratitis: Challenges in diagnosis and clinical management. Clin. Ophthalmol. 2017, 11, 185. [Google Scholar] [CrossRef] [PubMed]
  70. Bhela, S.; Mulik, S.; Gimenez, F.; Reddy, P.B.; Richardson, R.L.; Varanasi, S.K.; Jaggi, U.; Xu, J.; Lu, P.Y.; Rouse, B.T. Role of miR-155 in the pathogenesis of herpetic stromal keratitis. Am. J. Pathol. 2015, 185, 1073–1084. [Google Scholar] [CrossRef] [PubMed]
  71. Anand, S.; Majeti, B.K.; Acevedo, L.M.; Murphy, E.A.; Mukthavaram, R.; Scheppke, L.; Huang, M.; Shields, D.J.; Lindquist, J.N.; Lapinski, P.E. MicroRNA-132–mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nat. Med. 2010, 16, 909–914. [Google Scholar] [CrossRef]
  72. Suryawanshi, A.; Mulik, S.; Sharma, S.; Reddy, P.B.; Sehrawat, S.; Rouse, B.T. Ocular neovascularization caused by herpes simplex virus type 1 infection results from breakdown of binding betweenendothelial growth factor A and its soluble receptor. J. Immunol. 2011, 186, 3653–3665. [Google Scholar] [CrossRef] [PubMed]
  73. Yun, H.; Yee, M.B.; Lathrop, K.L.; Kinchington, P.R.; Hendricks, R.L.; Leger, A.J.S. Production of the cytokine VEGF-A by CD4+ T and myeloid cells disrupts the corneal nerve landscape and promotes herpes stromal keratitis. Immunity 2020, 53, 1050–1062.e5. [Google Scholar] [CrossRef] [PubMed]
  74. Mulik, S.; Xu, J.; Reddy, P.B.; Rajasagi, N.K.; Gimenez, F.; Sharma, S.; Lu, P.Y.; Rouse, B.T. Role of miR-132 in angiogenesis after ocular infection with herpes simplex virus. Am. J. Pathol. 2012, 181, 525–534. [Google Scholar] [CrossRef]
  75. Muraleedharan, C.K.; McClellan, S.A.; Barrett, R.P.; Li, C.; Montenegro, D.; Carion, T.; Berger, E.; Hazlett, L.D.; Xu, S. Inactivation of the miR-183/96/182 cluster decreases the severity of Pseudomonas aeruginosa-induced keratitis. Investig. Ophthalmol. Vis. Sci. 2016, 57, 1506–1517. [Google Scholar] [CrossRef] [PubMed]
  76. Coku, A.; McClellan, S.A.; Van Buren, E.; Back, J.B.; Hazlett, L.D.; Xu, S. The miR-183/96/182 cluster regulates the functions of corneal resident macrophages. ImmunoHorizons 2020, 4, 729–744. [Google Scholar] [CrossRef]
  77. Muraleedharan, C.K.; McClellan, S.A.; Ekanayaka, S.A.; Francis, R.; Zmejkoski, A.; Hazlett, L.D.; Xu, S. The miR-183/96/182 cluster regulates macrophage functions in response to Pseudomonas aeruginosa. J. Innate Immun. 2019, 11, 347–358. [Google Scholar] [CrossRef]
  78. Boomiraj, H.; Mohankumar, V.; Lalitha, P.; Devarajan, B. Human corneal microRNA expression profile in fungal keratitis. Investig. Ophthalmol. Vis. Sci. 2015, 56, 7939–7946. [Google Scholar] [CrossRef] [PubMed]
  79. Arenas-Padilla, M.; Mata-Haro, V. Regulation of TLR signaling pathways by microRNAs: Implications in inflammatory diseases. Cent. Eur. J. Immunol. 2018, 43, 482–489. [Google Scholar] [CrossRef] [PubMed]
  80. Cimmino, A.; Calin, G.A.; Fabbri, M.; Iorio, M.V.; Ferracin, M.; Shimizu, M.; Wojcik, S.E.; Aqeilan, R.I.; Zupo, S.; Dono, M. miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc. Natl. Acad. Sci. USA 2005, 102, 13944–13949. [Google Scholar] [CrossRef]
  81. Ahluwalia, J.K.; Khan, S.Z.; Soni, K.; Rawat, P.; Gupta, A.; Hariharan, M.; Scaria, V.; Lalwani, M.; Pillai, B.; Mitra, D. Human cellular microRNA hsa-miR-29a interferes with viral nef protein expression and HIV-1 replication. Retrovirology 2008, 5, 117. [Google Scholar] [CrossRef]
  82. Lobo, A.-M.; Agelidis, A.M.; Shukla, D. Pathogenesis of herpes simplex keratitis: The host cell response and ocular surface sequelae to infection and inflammation. Ocul. Surf. 2019, 17, 40–49. [Google Scholar] [CrossRef] [PubMed]
  83. Addis, H.; Jeng, B.H. Vernal keratoconjunctivitis. Clin. Ophthalmol. 2018, 12, 119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Arif, A.S.; Aaqil, B.; Siddiqui, A.; Nazneen, Z.; Farooq, U. Corneal complications and visual impairment in vernal keratoconjunctivitis patients. J. Ayub Med. Coll. Abbottabad 2017, 29, 58–60. [Google Scholar] [PubMed]
  85. Jing, L.; Li, H.; Zhang, T.; Lu, J.; Zhong, L. MicroRNA-4530 suppresses cell proliferation and induces apoptosis by targeting RASA1 in human umbilical vein endothelial cells. Mol. Med. Rep. 2019, 19, 3393–3402. [Google Scholar] [CrossRef]
  86. Pagliari, M.; Munari, F.; Toffoletto, M.; Lonardi, S.; Chemello, F.; Codolo, G.; Millino, C.; Della Bella, C.; Pacchioni, B.; Vermi, W. Helicobacter pylori affects the antigen presentation activity of macrophages modulating the expression of the immune receptor CD300E through miR-4270. Front. Immunol. 2017, 8, 1288. [Google Scholar] [CrossRef]
  87. Singh, N.; Diebold, Y.; Sahu, S.K.; Leonardi, A. Epithelial barrier dysfunction in ocular allergy. Allergy 2022, 77, 1360–1372. [Google Scholar] [CrossRef]
  88. Mantelli, F.; Mauris, J.; Argüeso, P. The ocular surface epithelial barrier and other mechanisms of mucosal protection: From allergy to infectious diseases. Curr. Opin. Allergy Clin. Immunol. 2013, 13, 563–568. [Google Scholar] [CrossRef]
  89. Lozano, V.; Martín, C.; Blanco, N.; Alcalde, I.; Fernandez-Vega Cueto, L.; Merayo-Lloves, J.; Quirós, L.M. Exosomes Released by Corneal Stromal Cells Show Molecular Alterations in Keratoconus Patients and Induce Different Cellular Behavior. Biomedicines 2022, 10, 2348. [Google Scholar] [CrossRef] [PubMed]
  90. Kwon, Y.H.; Fingert, J.H.; Kuehn, M.H.; Alward, W.L. Primary open-angle glaucoma. New Engl. J. Med. 2009, 360, 1113–1124. [Google Scholar] [CrossRef]
  91. Drewry, M.D.; Challa, P.; Kuchtey, J.G.; Navarro, I.; Helwa, I.; Hu, Y.; Mu, H.; Stamer, W.D.; Kuchtey, R.W.; Liu, Y. Differentially expressed microRNAs in the aqueous humor of patients with exfoliation glaucoma or primary open-angle glaucoma. Hum. Mol. Genet. 2018, 27, 1263–1275. [Google Scholar] [CrossRef]
  92. Sabirzhanov, B.; Zhao, Z.; Stoica, B.A.; Loane, D.J.; Wu, J.; Borroto, C.; Dorsey, S.G.; Faden, A.I. Downregulation of miR-23a and miR-27a following experimental traumatic brain injury induces neuronal cell death through activation of proapoptotic Bcl-2 proteins. J. Neurosci. 2014, 34, 10055–10071. [Google Scholar] [CrossRef] [PubMed]
  93. Jayaram, H.; Cepurna, W.O.; Johnson, E.C.; Morrison, J.C. MicroRNA expression in the glaucomatous retina. Investig. Ophthalmol. Vis. Sci. 2015, 56, 7971–7982. [Google Scholar] [CrossRef] [Green Version]
  94. Ren, C.; Hu, W.; Wei, Q.; Cai, W.; Jin, H.; Yu, D.; Liu, C.; Shen, T.; Zhu, M.; Liang, X. MicroRNA-27a Promotes Oxidative-Induced RPE Cell Death through Targeting FOXO1. BioMed Res. Int. 2021, 2021, 6666506. [Google Scholar] [CrossRef] [PubMed]
  95. Tabak, S.; Schreiber-Avissar, S.; Beit-Yannai, E. Crosstalk between MicroRNA and oxidative stress in primary open-angle glaucoma. Int. J. Mol. Sci. 2021, 22, 2421. [Google Scholar] [CrossRef] [PubMed]
  96. Parisi, C.; Napoli, G.; Amadio, S.; Spalloni, A.; Apolloni, S.; Longone, P.; Volonté, C. MicroRNA-125b regulates microglia activation and motor neuron death in ALS. Cell Death Differ. 2016, 23, 531–541. [Google Scholar] [CrossRef] [PubMed]
  97. Chen, H.; Cho, K.-S.; Vu, T.; Shen, C.-H.; Kaur, M.; Chen, G.; Mathew, R.; McHam, M.L.; Fazelat, A.; Lashkari, K. Commensal microflora-induced T cell responses mediate progressive neurodegeneration in glaucoma. Nat. Commun. 2018, 9, 3209. [Google Scholar] [CrossRef] [PubMed]
  98. Cowan, C.; Muraleedharan, C.K.; O’Donnell, J.J.; Singh, P.K.; Lum, H.; Kumar, A.; Xu, S. MicroRNA-146 inhibits thrombin-induced NF-κB activation and subsequent inflammatory responses in human retinal endothelial cells. Investig. Ophthalmol. Vis. Sci. 2014, 55, 4944–4951. [Google Scholar] [CrossRef]
  99. Zhuang, P.; Muraleedharan, C.K.; Xu, S. Intraocular delivery of miR-146 inhibits diabetes-induced retinal functional defects in diabetic rat model. Investig. Ophthalmol. Vis. Sci. 2017, 58, 1646–1655. [Google Scholar] [CrossRef]
  100. Comer, B.S.; Camoretti-Mercado, B.; Kogut, P.C.; Halayko, A.J.; Solway, J.; Gerthoffer, W.T. MicroRNA-146a and microRNA-146b expression and anti-inflammatory function in human airway smooth muscle. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2014, 307, L727–L734. [Google Scholar] [CrossRef]
  101. Boldin, M.P.; Taganov, K.D.; Rao, D.S.; Yang, L.; Zhao, J.L.; Kalwani, M.; Garcia-Flores, Y.; Luong, M.; Devrekanli, A.; Xu, J. miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice. J. Exp. Med. 2011, 208, 1189–1201. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, L.; Dong, L.; Tang, Y.; Li, M.; Zhang, M. MiR-146b protects against the inflammation injury in pediatric pneumonia through MyD88/NF-κB signaling pathway. Infect. Dis. 2020, 52, 23–32. [Google Scholar] [CrossRef]
  103. Kuchtey, J.; Rezaei, K.A.; Jaru-Ampornpan, P.; Sternberg, P.; Kuchtey, R.W. Multiplex cytokine analysis reveals elevated concentration of interleukin-8 in glaucomatous aqueous humor. Investig. Ophthalmol. Vis. Sci. 2010, 51, 6441–6447. [Google Scholar] [CrossRef] [Green Version]
  104. Zanon-Moreno, V.; Marco-Ventura, P.; Lleo-Perez, A.; Pons-Vazquez, S.; Garcia-Medina, J.J.; Vinuesa-Silva, I.; Moreno-Nadal, M.A.; Pinazo-Duran, M.D. Oxidative stress in primary open-angle glaucoma. J. Glaucoma 2008, 17, 263–268. [Google Scholar] [CrossRef] [PubMed]
  105. Baleriola, J.; García-Feijoo, J.; Martínez-de-la-Casa, J.M.; Fernández-Cruz, A.; Enrique, J.; Fernández-Durango, R. Apoptosis in the trabecular meshwork of glaucomatous patients. Mol. Vis. 2008, 14, 1513. [Google Scholar]
  106. Wang, L.; Wang, X.; Li, Z.; Kou, D.; Zhang, D.; Xu, Z. MiR-126 facilitates apoptosis of retinal ganglion cells in glaucoma rats via VEGF-Notch signaling pathway. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 8635–8641. [Google Scholar]
  107. Ye, P.; Liu, J.; He, F.; Xu, W.; Yao, K. Hypoxia-induced deregulation of miR-126 and its regulative effect on VEGF and MMP-9 expression. Int. J. Med. Sci. 2014, 11, 17. [Google Scholar] [CrossRef]
  108. Lang, G.E. Diabetic macular edema. Ophthalmologica 2012, 227, 21–29. [Google Scholar] [CrossRef] [PubMed]
  109. Yang, S.; Fei, X.; Lu, Y.; Xu, B.; Ma, Y.; Wan, H. miRNA-214 suppresses oxidative stress in diabetic nephropathy via the ROS/Akt/mTOR signaling pathway and uncoupling protein 2. Exp. Ther. Med. 2019, 17, 3530–3538. [Google Scholar] [CrossRef] [PubMed]
  110. Pang, B.; Qiao, L.; Wang, S.; Guo, X.; Xie, Y.; Han, L. MiR-214-3p plays a protective role in diabetic neuropathic rats by regulating Nav1. 3 and TLR4. Cell Biol. Int. 2021, 45, 2294–2303. [Google Scholar] [CrossRef] [PubMed]
  111. Yao, T.; Zha, D.; Gao, P.; Shui, H.; Wu, X. MiR-874 alleviates renal injury and inflammatory response in diabetic nephropathy through targeting toll-like receptor-4. J. Cell. Physiol. 2019, 234, 871–879. [Google Scholar] [CrossRef] [PubMed]
  112. Feng, B.; Chakrabarti, S. miR-320 regulates glucose-induced gene expression in diabetes. Int. Sch. Res. Not. 2012, 2012, 549875. [Google Scholar] [CrossRef] [PubMed]
  113. Aihara, M.; Kubota, N.; Minami, T.; Shirakawa, R.; Sakurai, Y.; Hayashi, T.; Iwamoto, M.; Takamoto, I.; Kubota, T.; Suzuki, R. Association between tear and blood glucose concentrations: Random intercept model adjusted with confounders in tear samples negative for occult blood. J. Diabetes Investig. 2021, 12, 266–276. [Google Scholar] [CrossRef] [PubMed]
  114. Wang, N.; Tan, H.-Y.; Feng, Y.-G.; Zhang, C.; Chen, F.; Feng, Y. microRNA-23a in human cancer: Its roles, mechanisms and therapeutic relevance. Cancers 2018, 11, 7. [Google Scholar] [CrossRef] [PubMed]
  115. Gupta, N.; Mansoor, S.; Sharma, A.; Sapkal, A.; Sheth, J.; Falatoonzadeh, P.; Kuppermann, B.; Kenney, M. Diabetic retinopathy and VEGF. Open Ophthalmol. J. 2013, 7, 4. [Google Scholar] [CrossRef] [Green Version]
  116. Nättinen, J.; Aapola, U.; Jylhä, A.; Vaajanen, A.; Uusitalo, H. Comparison of capillary and Schirmer strip tear fluid sampling methods using SWATH-MS proteomics approach. Transl. Vis. Sci. Technol. 2020, 9, 16. [Google Scholar] [CrossRef] [PubMed]
  117. Zhang, J.; Cui, C.; Xu, H. Downregulation of miR-145-5p elevates retinal ganglion cell survival to delay diabetic retinopathy progress by targeting FGF5. Biosci. Biotechnol. Biochem. 2019, 83, 1655–1662. [Google Scholar] [CrossRef] [PubMed]
  118. Xu, K.; Li, S.; Yang, Q.; Zhou, Z.; Fu, M.; Yang, X.; Hao, K.; Liu, Y.; Ji, H. MicroRNA-145-5p targeting of TRIM2 mediates the apoptosis of retinal ganglion cells via the PI3K/AKT signaling pathway in glaucoma. J. Gene Med. 2021, 23, e3378. [Google Scholar] [CrossRef]
  119. Hui, Y.; Yin, Y. MicroRNA-145 attenuates high glucose-induced oxidative stress and inflammation in retinal endothelial cells through regulating TLR4/NF-κB signaling. Life Sci. 2018, 207, 212–218. [Google Scholar] [CrossRef] [PubMed]
  120. Bresciani, E.; Saletti, C.; Squillace, N.; Rizzi, L.; Molteni, L.; Meanti, R.; Omeljaniuk, R.J.; Biagini, G.; Gori, A.; Locatelli, V. miRNA-218 targets lipin-1 and glucose transporter type 4 genes in 3T3-L1 cells treated with lopinavir/ritonavir. Front. Pharmacol. 2019, 10, 461. [Google Scholar] [CrossRef] [PubMed]
  121. Plaisance, V.; Abderrahmani, A.; Perret-Menoud, V.; Jacquemin, P.; Lemaigre, F.; Regazzi, R. MicroRNA-9 controls the expression of Granuphilin/Slp4 and the secretory response of insulin-producing cells. J. Biol. Chem. 2006, 281, 26932–26942. [Google Scholar] [CrossRef]
  122. Al-Muhtaresh, H.A.; Al-Kafaji, G. Evaluation of two-diabetes related microRNAs suitability as earlier blood biomarkers for detecting prediabetes and type 2 diabetes mellitus. J. Clin. Med. 2018, 7, 12. [Google Scholar] [CrossRef]
  123. Shen, J.; Yang, X.; Xie, B.; Chen, Y.; Swaim, M.; Hackett, S.F.; Campochiaro, P.A. MicroRNAs regulate ocular neovascularization. Mol. Ther. 2008, 16, 1208–1216. [Google Scholar] [CrossRef]
  124. Wang, S.; Li, K. MicroRNA-96 regulates RGC-5 cell growth through caspase-dependent apoptosis. Int. J. Clin. Exp. Med. 2014, 7, 3694. [Google Scholar]
  125. Kumar, A.; Sidhu, J.; Goyal, A.; Tsao, J.W.; Svercauski, J. Alzheimer disease (nursing). Health Aff. 2021, 33, 650–657. [Google Scholar]
  126. Hébert, S.S.; Papadopoulou, A.S.; Smith, P.; Galas, M.-C.; Planel, E.; Silahtaroglu, A.N.; Sergeant, N.; Buée, L.; De Strooper, B. Genetic ablation of Dicer in adult forebrain neurons results in abnormal tau hyperphosphorylation and neurodegeneration. Hum. Mol. Genet. 2010, 19, 3959–3969. [Google Scholar] [CrossRef] [PubMed]
  127. Kalló, G.; Emri, M.; Varga, Z.; Ujhelyi, B.; Tőzsér, J.; Csutak, A.; Csősz, É. Changes in the chemical barrier composition of tears in Alzheimer’s disease reveal potential tear diagnostic biomarkers. PLoS ONE 2016, 11, e0158000. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Higaki, S.; Muramatsu, M.; Matsuda, A.; Matsumoto, K.; Satoh, J.-I.; Michikawa, M.; Niida, S. Defensive effect of microRNA-200b/c against amyloid-beta peptide-induced toxicity in Alzheimer’s disease models. PLoS ONE 2018, 13, e0196929. [Google Scholar] [CrossRef] [PubMed]
  129. Liu, C.-G.; Wang, J.-L.; Li, L.; Wang, P.-C. MicroRNA-384 regulates both amyloid precursor protein and β-secretase expression and is a potential biomarker for Alzheimer’s disease. Int. J. Mol. Med. 2014, 34, 160–166. [Google Scholar] [CrossRef] [PubMed]
  130. Scully, O.J.; Bay, B.-H.; Yip, G.; Yu, Y. Breast cancer metastasis. Cancer Genom. Proteom. 2012, 9, 311–320. [Google Scholar]
  131. Lim, B.; Hortobagyi, G.N. Current challenges of metastatic breast cancer. Cancer Metastasis Rev. 2016, 35, 495–514. [Google Scholar] [CrossRef] [PubMed]
  132. Loh, H.-Y.; Norman, B.P.; Lai, K.-S.; Rahman, N.M.A.N.A.; Alitheen, N.B.M.; Osman, M.A. The regulatory role of microRNAs in breast cancer. Int. J. Mol. Sci. 2019, 20, 4940. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, G.; Zhang, W.; Li, B.; Stringer-Reasor, E.; Chu, C.; Sun, L.; Bae, S.; Chen, D.; Wei, S.; Jiao, K. MicroRNA-200c and microRNA-141 are regulated by a FOXP3-KAT2B axis and associated with tumor metastasis in breast cancer. Breast Cancer Res. 2017, 19, 73. [Google Scholar] [CrossRef] [PubMed]
  134. Yan, L.-X.; Huang, X.-F.; Shao, Q.; Huang, M.-Y.; Deng, L.; Wu, Q.-L.; Zeng, Y.-X.; Shao, J.-Y. MicroRNA miR-21 overexpression in human breast cancer is associated with advanced clinical stage, lymph node metastasis and patient poor prognosis. RNA 2008, 14, 2348–2360. [Google Scholar] [CrossRef] [PubMed]
  135. Volinia, S.; Calin, G.A.; Liu, C.-G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261. [Google Scholar] [CrossRef]
  136. Feng, Y.-H.; Tsao, C.-J. Emerging role of microRNA-21 in cancer. Biomed. Rep. 2016, 5, 395–402. [Google Scholar] [CrossRef] [PubMed]
  137. Chan, J.A.; Krichevsky, A.M.; Kosik, K.S. MicroRNA-21 is an antiapoptotic factor in human glioblastoma cells. Cancer Res. 2005, 65, 6029–6033. [Google Scholar] [CrossRef]
  138. Löffler, D.; Brocke-Heidrich, K.; Pfeifer, G.; Stocsits, C.; Hackermüller, J.; Kretzschmar, A.K.; Burger, R.; Gramatzki, M.; Blumert, C.; Bauer, K. Interleukin-6–dependent survival of multiple myeloma cells involves the Stat3-mediated induction of microRNA-21 through a highly conserved enhancer. Blood J. Am. Soc. Hematol. 2007, 110, 1330–1333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Zhu, S.; Wu, H.; Wu, F.; Nie, D.; Sheng, S.; Mo, Y.-Y. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res. 2008, 18, 350–359. [Google Scholar] [CrossRef]
  140. Toiyama, Y.; Hur, K.; Tanaka, K.; Inoue, Y.; Kusunoki, M.; Boland, C.R.; Goel, A. Serum miR-200c is a novel prognostic and metastasis-predictive biomarker in patients with colorectal cancer. Ann. Surg. 2014, 259, 735. [Google Scholar] [CrossRef]
  141. Jurmeister, S.; Baumann, M.; Balwierz, A.; Keklikoglou, I.; Ward, A.; Uhlmann, S.; Zhang, J.D.; Wiemann, S.; Sahin, Ö. MicroRNA-200c represses migration and invasion of breast cancer cells by targeting actin-regulatory proteins FHOD1 and PPM1F. Mol. Cell. Biol. 2012, 32, 633–651. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Overall pipeline for the discovery of tear film miRNAs as biomarkers.
Figure 1. Overall pipeline for the discovery of tear film miRNAs as biomarkers.
Ijms 24 03694 g001
Table 1. Summary of literature reporting tear microRNAs as potential biomarkers.
Table 1. Summary of literature reporting tear microRNAs as potential biomarkers.
StudyDiseaseNumber of ParticipantsTear Collection MethodDetection Method
Wang, Q. et al. (2020) [21]Dry eye Disease (DED)138 HC; 138 DEDCapillary tubemiRNA microarray
Pucker, A.D. et al. (2022) [29] Dry Eye Disease (DED)5 HC; 5 DEDPhosphate buffer saline (PBS) washRNA-Seq
Kim, Y.J. et al. (2019) [19]Sjögren Syndrome (SS)8 HC; 18 SSMicropipettesRT-PCR
Kim, Y.J. et al. (2022) [22]Herpes Epithelial Keratitis (HEK)7 HC; 8 HEKMicropipettesRT-PCR
Syed, N.H. et al. (2022) [31]Vernal Keratoconjunctivitis (VKC)4 HC; 4 VKCSchirmer stripmiRNA microarray
Tamkovich, S. et al. (2019) [28]Primary Open Angle Glaucoma (POAG)29 HC; 33 POAGCapillary tubeRT-PCR
Raga-Cervera, J. et al. (2021) [20]Primary Open Angle Glaucoma (POAG)22 OHT; 20 POAGCapillary tubeRNA-Seq
Chan, H.W. et al. (2020) [12] Diabetic Macular Edema (DME)24 DMESchirmer stripRT-PCR
Sun, L. et al. (2021) [18]Diabetic Retinopathy (DR)28 HC; 33 DRCapillary tubeRT-PCR
Hu, L. et al. (2022) [27]Diabetic Retinopathy (DR)11 HC; 10 DM; 9 DRSchirmer stripRNA-Seq
Kenny, A. et al. (2019) [16]Alzheimer’s Disease (AD)15 HC; 8 MCI; 9 ADSchirmer stripRT-PCR
Inubushi, S. et al. (2020) [32]Metastatic Breast Cancer (MBC)8 HC; 5 MBCSchirmer stripRT-PCR
HC = Healthy control; OHT = Ocular hypertension; DM = Diabetes mellitus; MCI = mild cognitive impairment.
Table 2. Up and down-regulated tear microRNAs reported as potential biomarkers.
Table 2. Up and down-regulated tear microRNAs reported as potential biomarkers.
ConditionTear miRNAs in Disease State vs. Controls
UpregulatedDownregulated
Dry Eye Disease miR-127-5p, miR-1273h-3p, miR-1288-5p, miR-130b-5p, miR-139-3p, miR-1910-5p, miR-203b-5p, miR-22-5p, miR-4632-3p, miR-450b-5p, miR-1283, miR-5700, miR-3671 miR-4673, miR-890, miR-576-5p, miR-337-3p, miR-607, miR-1827, miR-485-5p, miR-6884-5p, miR-335-3p, miR-647, miR-4433b-5p, miR-570-3p, miR-1276, miR-2054, miR-802, miR-539-5p, miR-198, miR-3646, miR-4427, miR-4705, miR-606, miR-20b-3p, miR-4475, miR-4478, miR-4797-5p, miR-142-5p, miR-153-5p, miR-5583-5p
Sjögren’s SyndromemiR-16-5p, miR-34a-5p, miR-142-3p, miR-223-3pmiR-30b-5p, miR-30c-5p, miR-30d-5p, miR-92a-3p, miR-134-5p, miR-137, miR-302d-5p, miR-365b-3p, miR-374c-5p, miR-487b-3p
Herpes Epithelial KeratitismiR-15b-5p, miR-16-5p, miR-20b-5p, miR-21-5p, miR-23b-3p, miR-25-3p, miR-29a-3p, miR-30a-3p, miR-30d-5p, miR-92a-3p, miR-124-3p, miR-127-3p, miR-132-3p, miR-142-3p, miR-145-5p, miR-146a-5p, miR-146b-5p, miR-155-5p, miR-182-5p, miR-183-5p, miR-221-3p, miR-223-3p, miR-338-5p
Vernal KeratoconjunctivitismiR-1229-5p, miR-6821-5p, miR-6800-5p, miR-4466, miR-3665, miR-4530, miR-7110-5p, miR-1207-5p, miR-6875-5p, miR-762, miR-4741, miR-6740-5p, miR-4298, miR-7107-5p, miR-2861, miR-3663-3p, miR-6891-5p, miR-4672, miR-6785-5p, miR-6510-5p, miR-6803-5p, miR-718, miR-642b-3p, miR-6124, miR-4687-3p, miR-4721, miR-4459, miR-8072, miR-5703, miR-3195, miR-7847-3p, miR-4665-3p, miR-6869-5p, miR-638, miR-6087, miR-4516, miR-3960, miR-6089, miR-4443, miR-5787, miR-642a-3p, miR-3679-5p, miR-6088, miR-8069, miR-4281, miR-1273g-3p, miR-7150, miR-940miR-7975, miR-7977, miR-1260a
Primary Open-Angle GlaucomamiR-26b-5p, miR-27a-3p, miR-152-3p, miR-30e-5p, miR-125b-2-5p, miR-224-5pmiR-151a-3p, miR-1307-3p, miR-146b, miR-16, miR-126
Response to Anti-VEGF Treatments for Diabetic Macular EdemaCorrelated with good response: miR-214-3p, miR-320d, miR-874-3p, Correlated with poor response: miR-98-5p, miR-196b-5p, miR-454-3p
Diabetic RetinopathymiR-9-5p, miR-143-3p, miR-411-5p, miR-218-5p, miR-139-5p, miR-10b-5p, miR-129-5p, miR-214-3p, miR-381-3p, miR-130b-4p, miR-382-5p, miR-145-5p, miR-379-5p, miR-127-3p, miR-196a-5p, miR-206, miR-1-3p, miR-199a-5p, miR-642a-3p, miR-642b-5pmiR-23a, miR-361-3p, miR-29c-5p, miR-532-5p, miR-3615, miR-429, miR-224-5p, miR-146a-5p, miR-149-5p, miR-339-3p, miR-203a-3p, miR-24-1-5p, miR-203b-5p, miR-31-5p, miR-147b-3p, miR-205-5p, miR-186-5p, miR-7706, miR-191-5p, miR-135b-5p, miR-96-5p
Alzheimer’s DiseasemiR-200b-5p
Metastatic Breast CancermiR-21, miR-200c
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

Altman, J.; Jones, G.; Ahmed, S.; Sharma, S.; Sharma, A. Tear Film MicroRNAs as Potential Biomarkers: A Review. Int. J. Mol. Sci. 2023, 24, 3694. https://doi.org/10.3390/ijms24043694

AMA Style

Altman J, Jones G, Ahmed S, Sharma S, Sharma A. Tear Film MicroRNAs as Potential Biomarkers: A Review. International Journal of Molecular Sciences. 2023; 24(4):3694. https://doi.org/10.3390/ijms24043694

Chicago/Turabian Style

Altman, Jeremy, Garrett Jones, Saleh Ahmed, Shruti Sharma, and Ashok Sharma. 2023. "Tear Film MicroRNAs as Potential Biomarkers: A Review" International Journal of Molecular Sciences 24, no. 4: 3694. https://doi.org/10.3390/ijms24043694

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