Abstract
Purpose:
Diabetic keratopathy (DK) is a vision-threatening disease that occurs in people with diabetes. Mounting evidence indicates that microRNAs (miRNAs) are indispensable in nerve regeneration within DK. Herein, the role of miRNAs associated with DK, especially focusing on autophagy and apoptosis regulation, was investigated.
Methods:
To identify differentially expressed miRNAs, we performed miRNA sequencing on trigeminal ganglion (TG) tissues derived from streptozotocin-induced type 1 diabetic mellitus (T1DM) and normal mice. MiR-144-3p was chosen for the subsequent experiments. To explore the regulatory role of miR-144-3p in DK, miRNA antagomir was utilized to inhibit miR-144-3p expression. Bioinformatic tools were used to predict the target genes of miR-144-3p, and a dual-luciferase reporter assay was then applied for validation. Autophagy and apoptosis activities were measured utilizing TUNEL staining, immunofluorescence staining, and Western blotting.
Results:
Overall, 56 differentially expressed miRNAs were detected in diabetic versus control mice. In the diabetic mouse TG tissue, miR-144-3p expression was aberrantly enhanced, whereas decreasing its expression contributed to improved diabetic corneal re-epithelialization and nerve regeneration. Fork-head Box O1 (FOXO1) was validated as a target gene of miR-144-3p. Overexpression of FOXO1 could prevent both inadequate autophagy and excessive apoptosis in DK. Consistently, a specific miR-144-3p inhibition enhanced autophagy and prevented apoptosis in DK.
Conclusions:
In this study, our research confirmed the target binding relationship between miR-144-3p and FOXO1. Inhibiting miR-144-3p might modulate autophagy and apoptosis, which could generate positive outcomes for corneal nerves via targeting FOXO1 in DK.
Diabetes mellitus (DM) is a metabolic disease featured by long-lasting hyperglycemia that can cause nerve damage in multiple organs. The Diabetes Atlas, launched in 2021, hints that over 500 million adults suffer from diabetes globally.
1 Among the various complications caused by DM, diabetic keratopathy (DK) is the leading ocular disorder.
2 Neurons originating from ophthalmic sections of the trigeminal nerve constitute most corneal nerve fibers.
3 These corneal nerve fibers cooperate with epithelial cells to generate neuropeptides and neurotrophic factors and maintain the homeostasis of the cornea.
4,5 Clinically, DK is characterized by changes in tear film, delayed epithelial regeneration, attenuative sensitivity, and corneal ulceration, which might ultimately lead to sight-threatening consequences.
6 Recent studies indicate the involvement of DNA methylation, microRNA (miRNA) action, and sympathetic nerve overactivation in the corneal epithelial wound healing process in DK.
3,7 Moreover, dysregulation of growth factor, neurotrophin, inflammation, and autophagy signaling pathways is considered to be related to the pathogenesis of DK.
8,9 However, even if there is a pressing need for effective therapeutic interventions to promote corneal nerve regeneration, the existing treatments have limited effects, and the pathogenic pathways involved in DK remain unclear.
10 The necessity of elucidating the mechanism and searching for potential targets in DK served as the impetus for this work.
Through microarray analysis and high-throughput sequencing, novel regulatory and biomarker molecules for various diseases could be identified effectively. In terms of neurodegenerative disorders, machine learning techniques and high-throughput sequencing have identified 90 independent risk-associated genetic variants in Parkinson's disease.
11 In addition, the matrix metalloproteinase-12 was shown to upregulate in diabetic peripheral neuropathy by microarray and protein alteration assessment in type 2 diabetic mice.
12 In mapping the anterior segment cell types of the human eye, single-nucleus RNA sequencing was combined with Mendelian inheritance patterns to reveal over 900 human ocular disease genes.
13 Moreover, the microarray analysis was also utilized to identify 29 differentially expressed miRNAs (DEmiRNAs) in the human diabetic and normal corneas.
14
Our previous work showed that some specific noncoding RNAs (ncRNAs) can regulate diabetic corneal disease in different ways.
15–17 Among them, miRNAs are evolutionarily conserved and act as regulators of various signaling pathways by inducing mRNA degradation or suppressing translational progress.
18 For example, miR-34c and miR-146a were found to affect nerve recovery and inflammatory responses during diabetic corneal epithelial wound healing.
15,19 In addition, miR-223-5p was shown to contribute to corneal epithelial and nerve healing through modulating inflammation and proliferation under hyperglycemic conditions.
20 The long noncoding RNA Rik was proven to be a competitive endogenous RNA (ceRNA) that can bind to miR-181a-5p and improve diabetic corneal epithelial wound healing.
21 These studies consistently report the presence of regulatory miRNAs in DK and the necessity of further study.
In the present study, we combined next-generation sequencing and bioinformatic analyses of DEmiRNAs obtained from the trigeminal ganglion (TG) tissues of normal mice and diabetic mice to efficiently identify novel targets. After preliminary validation, the miR-144-3p/Fork-head Box O1 (FOXO1) axis was determined for further investigation. Our data reveal that inhibition of miR-144-3p might improve re-epithelialization and nerve regeneration in the diabetic cornea by promoting autophagy and suppressing apoptosis via FOXO1.
Male C57BL/6 mice (6-8 weeks old) were purchased from SPF Biotechnology Co., Ltd. (Beijing, China), and resided in the Fujian Medical University Laboratory Animal Center. All animal studies were approved by the Biomedical Research Ethics Review Committee of Fujian Medical University (IACUC FJMU 2021-0454). Establishment of the type 1 diabetic mouse model was conducted by injecting streptozotocin (STZ, 50 mg/kg; Sigma-Aldrich, St. Louis, MO, USA) intraperitoneally for 5 successive days. Blood glucose levels in the tail veins and body weight were assessed. Mice with blood glucose levels exceeding 16.67 mmol/L after 16 weeks of STZ injection were identified as diabetic mice and used in the subsequent study.
We collected the TG tissues of mice and cut them into pieces. Total mRNA and miRNA were obtained by TRIzol reagent (Invitrogen, Waltham, MA, USA). Complementary deoxyribonucleic acids (cDNAs) were synthesized by Thermo Scientific Revertaid First Strand cDNA Synthesis kit (Waltham, MA, USA). Quantitative real-time PCR (qRT-PCR) was performed by 2 × PCR master mix (Arraystar, Rockville, MD, USA). The β-actin gene and the U6 gene were utilized as an internal control.
We collected TG tissues from mice and lysed them with RIPA reagent in order to extract the proteins. Sample proteins were run on SDS-PAGE and transferred using PVDF membranes. Then, 5% skimmed milk was applied to the block. Primary antibodies include anti-FOXO1 (ab179450; Abcam, Waltham, MA, USA), anti-P62 (P0067; Sigma), anti-LC3B (ab192890; Abcam), anti-BCL2 (ab182858; Abcam), and β-actin (Sigma). Membranes were then incubated with corresponding antibodies overnight. The next day, we incubated membranes with the secondary antibodies horseradish peroxidase (HRP; ab6721; Abcam) for 1 hour after rinsing the membranes with TBS + Tween (TBST) thrice. Finally, enhanced chemiluminescence (ECL) solution (Millipore, St. Louis, MO, USA) was utilized to visualize the protein bands.
TG tissues of mice were fixed in optimum cutting temperature (OCT) compound (Tissue-Tek, Torrance, CA, USA). Seven-micrometer frozen sections were prepared. The samples were then fixed in 4% paraformaldehyde, permeabilized with 0.3% Triton X-100, blocked with 5% BSA, and incubated with primary antibody. The antibodies were as follows: anti-FOXO1 (ab179450; Abcam), anti-P62 (P0067; Sigma), anti-LC3B (ab192890; Abcam), and anti- BCL2 (ab182858; Abcam). Sections were then incubated with Alexa Fluor–conjugated secondary antibodies (Proteintech, Wuhan, China). Finally, after counterstaining with DAPI (Beyotime, China), the stained sections were visualized under a DMI8 inverted fluorescence microscope (Leica, Wetzlar, Germany).
Hyperglycemia Impairs Corneal Re-Epithelialization and Nerve Regeneration in Mice
Bioinformatics Analyses of Differentially Expressed miRNAs in Diabetic TG Tissues
Inhibition of miR-144-3p Ameliorated Diabetic Corneal Re-Epithelial and Nerve Regeneration
Diabetic Mice Displayed Defective Autophagy and Excessive Apoptosis in TG Tissues
Overexpression of FOXO1 Promoted Autophagy and Reduced Apoptosis in Diabetic TG Tissues
Suppressing miR-144-3p Promoted Autophagy and Reduced Apoptosis in Diabetic TG Tissues