March 2024
Volume 65, Issue 3
Open Access
Cornea  |   March 2024
NEAT1 Deficiency Promotes Corneal Epithelial Wound Healing by Activating cAMP Signaling Pathway
Author Affiliations & Notes
  • Tian Sang
    State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
  • Yani Wang
    State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
  • Zhiqing Wang
    School of Clinical Medicine, Weifang Medical University, Shandong, China
  • Di Sun
    State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
  • Shengqian Dou
    State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
  • Yaoyao Yu
    State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
  • Xiaoyun Wang
    State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
  • Can Zhao
    Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
    School of Ophthalmology, Shandong First Medical University, Shandong, China
  • Qun Wang
    State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
  • Correspondence: Qun Wang, 5 Yan'erdao Road, Qing dao, Shandong 266071, China; [email protected]
  • Can Zhao, 372 Jingsi Road, Jinan, Shandong 250021, China; [email protected]
Investigative Ophthalmology & Visual Science March 2024, Vol.65, 10. doi:https://doi.org/10.1167/iovs.65.3.10
  • Views
  • PDF
  • Share
  • Tools
    • Alerts
      ×
      This feature is available to authenticated users only.
      Sign In or Create an Account ×
    • Get Citation

      Tian Sang, Yani Wang, Zhiqing Wang, Di Sun, Shengqian Dou, Yaoyao Yu, Xiaoyun Wang, Can Zhao, Qun Wang; NEAT1 Deficiency Promotes Corneal Epithelial Wound Healing by Activating cAMP Signaling Pathway. Invest. Ophthalmol. Vis. Sci. 2024;65(3):10. https://doi.org/10.1167/iovs.65.3.10.

      Download citation file:


      © ARVO (1962-2015); The Authors (2016-present)

      ×
  • Supplements
Abstract

Purpose: This study aimed to investigate the role of the long non-coding RNA (lncRNA) NEAT1 in corneal epithelial wound healing in mice.

Methods: The central corneal epithelium of wild-type (WT), MALAT1 knockout (M-KO), NEAT1 knockout (N-KO), and NEAT1 knockdown (N-KD) mice was scraped to evaluate corneal epithelial and nerve regeneration rates. RNA sequencing of the corneal epithelium from WT and N-KO mice was performed 24 hours after debridement to determine the role of NEAT1. Quantitative PCR (qPCR) and ELISA were used to confirm the bioinformatic analysis. The effects of the cAMP signaling pathway were evaluated in N-KO and N-KD mice using SQ22536, an adenylate cyclase inhibitor.

Results: Central corneal epithelial debridement in N-KO mice significantly promoted epithelial and nerve regeneration rates while suppressing inflammatory cell infiltration. Furthermore, the expression of Atp1a2, Ppp1r1b, Calm4, and Cngb1, which are key components of the cAMP signaling pathway, was upregulated in N-KO mice, indicative of its activation. Furthermore, the cAMP pathway inhibitor SQ22536 reversed the accelerated corneal epithelial wound healing in both N-KO and N-KD mice.

Conclusions: NEAT1 deficiency contributes to epithelial repair during corneal wound healing by activating the cAMP signaling pathway, thereby highlighting a potential therapeutic strategy for corneal epithelial diseases.

The cornea is a transparent avascular tissue that serves as the primary structural barrier to the remaining ocular tissue. The cornea consists of three main layers: corneal epithelium, stroma, and endothelium.1 The corneal epithelium, which is the outer surface of the eye, is susceptible to physical, chemical, and infectious insults that often result in corneal epithelial damage. Corneal epithelial wounds typically heal rapidly and effectively within a few days, restoring structural and functional integrity.24 Corneal epithelial regeneration is a complex and dynamic process that involves the repair of the epithelial layer, cell migration, and proliferation during wound healing.57 However, untreated corneal epithelial injury can lead to complications, such as inflammation, neovascularization, ulceration, scarring, and other complications, significantly impairing vision.8 
Long non-coding RNAs (lncRNAs) are transcripts that are at least 200 nucleotides in length and possess limited protein-encoding ability.9 Due to a higher total quantity than coding genes,10 the diversity and size of lncRNAs are strongly correlated with organismal complexity, surpassing the correlation observed for protein-coding genes.11,12 Aberrant lncRNA expression has been implicated in various human diseases, including tumorigenesis, neurological diseases, and cardiovascular diseases.13 LncRNAs have been extensively studied in the field of ophthalmology, particularly in relation to Fuchs’ endothelial corneal dystrophy (FECD), glaucoma, cataracts, retinal disease, and ocular tumors.1418 Among these lncRNAs, nuclear paraspeckle assembly transcript 1 (NEAT1) has shown promise in its involvement in various ocular diseases and its ability to exert functional effects.19,20 Previous studies have indicated that the downregulation of NEAT1 expression inhibits neovascularization of the cornea,21 whereas NEAT1 deficiency impairs the oxidant–antioxidant balance in the corneal endothelium.14 In the corneal epithelium, NEAT1 accelerating diabetes-related dry eye disease development and NEAT1 knockdown will alleviate corneal damage.22 Moreover, NEAT1 plays a role in abnormal immune activation and is related to the disease progression of primary Sjögren’s syndrome, affecting the health of the corneal epithelium.23 These studies suggest that the expression of NEAT1 in the cornea maintains a balance, and excessive or insufficient expression affects the corneal homeostasis. Such studies also indicate diverse functions of NEAT1 across different cell types. However, the role of NEAT1 in protecting corneal epithelium during wound healing remains unclear. 
This study aimed to delineate the role of NEAT1 in corneal epithelial wound healing in mice. The findings show that the regeneration of both epithelial and nerve tissues was significantly enhanced in NEAT1 knockout (N-KO) mice, whereas inflammatory cell infiltration was reduced. The cAMP signaling pathway was significantly activated in N-KO mice, which was confirmed by the upregulated expression of Atp1a2, Ppp1r1b, Calm4, Cngb1, and cAMP production in the corneal epithelium. Additionally, administration of the cAMP pathway inhibitor SQ22536 reversed the accelerated healing of corneal epithelial wounds in N-KO and NEAT1 knockdown (N-KD) mice. In conclusion, our data substantiate the role of NEAT1 in corneal epithelial wound healing and suggest its potential as a therapeutic target for corneal epithelial diseases. 
Methods
Animals
Male C57BL/6 mice (8 weeks old) were procured from Beijing Vital River Laboratory Animal Technology (Beijing, China). N-KO mice (strain number T011757) were purchased from GemPharmatech (Nanjing, China) and bred as heterozygotes in a specific pathogen-free environment. Homozygous mice derived from heterozygote breeding were used in the present study. 
Recombinant adeno-associated virus (AAV) was constructed using inverted terminal repeats from serotype 2 AAV and capsid protein from serotype 9. AAV containing NEAT1 short hairpin RNA (shRNA) and control shRNA (NC) target sequences were obtained from GeneChem (Shanghai, China). All of the sequences are shown in Table 1. Following intraperitoneal injection of 0.6% pentobarbital sodium (50 mg/kg) and the application of topical anesthesia (2% xylocaine), the male C57BL/6 mice were administered the AAV virus by subconjunctival injection (at ∼5 × 1012 vector genomes per milliliter, 5 µL per eye) to generate N-KD mice. RNA was extracted from mouse corneal epithelium after at least 4 weeks to detect infection efficiency. 
Table 1.
 
Target Sequences
Table 1.
 
Target Sequences
The 200-µM cAMP inhibitor SQ22536 (Selleck Chemicals, Houston, TX, USA) was administered by subconjunctival injection. Mice were systemically anesthetized with pentobarbital sodium (50 mg/kg, intraperitoneally) and topically anesthetized with 2% xylocaine. SQ22536 (5 µL per eye) was injected 24 hours before, at 0 hour, and at 24 hours after wounding. cAMP production of epithelium after SQ22536 treatment was determined to detect an inhibitory effect. 
The mice were housed at the animal center of the Shandong Eye Institute, where they had unrestricted access to food and water. All protocols involving animals were approved by the Ethics Committee of Shandong Eye Institute and conducted in accordance with the guidelines outlined in the ARVO Statement for the Use of Animals in Ophthalmic and Vision Research. 
Data Processing and Downstream Analysis of Single-Cell RNA Sequencing
We employed the Cell Ranger 3.1.0 pipeline to map the cleaned RNA sequencing (RNA-seq) reads to the human reference genome (GRCh38-3.0.0) based on the early sequencing results (databases hra00781 and hra00728). The count data were subsequently imported into the Seurat 3.2.0 R package (R Foundation for Statistical Computing, Vienna, Austria).24 The raw reads underwent processing, which involved the removal of adaptor sequences, short sequences, low-quality bases, and ambiguous sequences (reads with more than two unknown bases N). Subsequently, the canonical correlation analysis algorithm was used to exclude batch effects during data integration,25 and the data were normalized using the Seurat LogNormalize function. To complete cell clustering, principal component analysis was performed for highly variable genes. Genes selected for this analysis underwent visualized cell clustering using Uniform Manifold Approximation and Projection (UMAP) stochastic neighbor embedding to generate four cluster types. 
Corneal Epithelial Wound Healing
To establish the corneal epithelial debridement model, mice were anesthetized with intraperitoneal injections of 0.6% pentobarbital sodium (50 mg/kg), followed by topical administration of 2% xylocaine. The central corneal epithelium (2.5 mm in diameter) was excised using AlgerBrush II Corneal Rust Ring Remover (Alger, Lago Vista, TX, USA).26 The procedure was performed to prevent infection, and ofloxacin eye drops were administered after debridement. Corneal epithelial defects were evaluated at 0, 12, 24, and 36 hours after debridement using 0.25% fluorescein sodium and were visualized using a slit-lamp microscope. The defect area was quantified using ImageJ software (National Institutes of Health, Bethesda, MD, USA).27 
Hematoxylin and Eosin Staining
Eyeballs were collected after a 24-hour interval following the induction of corneal epithelial injury. The collected eyeballs were fixed overnight with tissue fixative. Subsequently, the samples were dehydrated, paraffin embedded, and sectioned at a 5-µm thickness. The sections subsequently underwent hematoxylin and eosin (H&E) staining with a diaminobenzidine chromogenic solution. 
Quantitative Real-Time PCR
Mouse eyeballs were placed in Dulbecco's Modified Eagle Medium/Nutrient Mixture F-12 (DMEM/F-12; Wuhan Pricella Biotechnology, Wuhan, China) containing 15 mg/mL Dispase II (Roche, Mannheim, Germany) and incubated overnight at 4°C. Subsequently, the epithelium was delicately detached using a dissecting microscope.28 Total RNA was extracted from corneal epithelial tissue using the TransZol Up Plus RNA Kit (TransGen Biotech, Beijing, China), and cDNA was synthesized using a reverse transcription kit (Vazyme Biotech, Nanjing, China). Subsequently, quantitative PCR (qPCR) was conducted employing the ChamQ Universal SYBR qPCR Master Mix (Vazyme Biotech) and a quantitative real-time PCR (qRT-PCR) instrument (Applied Biosystems, Carlsbad, CA, USA). Each experiment was performed in triplicate. The cycling conditions for qPCR consisted of an initial step at 95°C for 30 seconds followed by 40 two-step cycles of 10 seconds at 95°C and 30 seconds at 60°C. β-Actin served as the internal control for messenger RNA (mRNA) and lncRNA. The primer pairs (Delohaida, Qingdao, China) used for the qRT-PCR analyses are listed in Table 2
Table 2.
 
Primer Pairs Used for qRT-PCR Analyses
Table 2.
 
Primer Pairs Used for qRT-PCR Analyses
Corneal Whole-Mount Staining for Nerve Fibers
Mouse eyeballs were immersed in Zamboni fixative (Beijing Solarbio Science & Technology, Beijing, China) for 1 hour 5 days following corneal epithelial injury. Subsequently, the cornea was completely separated and immersed in PBS containing 0.3% Triton X-100 and 3% BSA. The samples were sealed airtight and left overnight at 4°C. Next, the cornea was tagged with an Alexa Fluor 647 anti-Tubulin-β III Antibody (1:400; BioLegend, San Diego, CA, USA) and incubated overnight at 4°C. After each cornea was thoroughly rinsed six times with PBS containing 0.3% Triton X-100 and 0.05% Tween 20, the cornea was divided into six segments or “petals.” These segments were observed under a confocal microscope.29 Corneal nerve fiber density was quantified using ImageJ. 
RNA Sequencing and Data Processing
Thirty-two regenerative corneal epithelium samples were collected from equal numbers of wild-type (WT) and N-KO mice. The samples were collected 24 hours after debridement and were subsequently combined to form eight distinct groups, with four groups consisting of normal mice with corneal injury and four groups comprised of N-KO mice with corneal injury. Each group consisted of corneal epithelial samples obtained from four mice. The collected epithelial samples were stored at −80°C for the subsequent experimental procedures. 
Total RNA was extracted using Invitrogen TRIzol Reagent (Thermo Fisher Scientific, Waltham, MA, USA). Subsequently, the libraries were constructed utilizing the VAHTS Universal V6 RNA-seq Library Prep Kit for Illumina (Vazyme Biotech). Transcriptome sequencing and analysis were conducted by Shanghai OE Biotech (Shanghai, China). The libraries were sequenced on an Illumina NovaSeq 6000 platform (Illumina, San Diego, CA, USA). The obtained raw reads in fastq format were first processed using fastp,30 and the low quality reads were then removed to obtain the clean reads. These clean reads were mapped to the reference genome using HISAT2.31 The fragments per kilobase per million mapped fragments (FPKM)32 of each gene were calculated, and the read counts of each gene were determined with HTSeq-count.33 Principal component analysis was conducted using R 3.2.0 to evaluate the biological duplication of samples. DESeq234 was employed for differential expression analysis. Q < 0.05 and fold change > 2 or fold change < 0.5 were set as the thresholds for significantly differential expression genes (DEGs). Based on the hypergeometric distribution, Kyoto Encyclopedia of Genes and Genomes (KEGG)35 pathway enrichment analyses of DEGs were performed to screen the significant enriched term using R 3.2.0. 
ELISA Analysis
Mouse corneal epithelium was collected 48 hours post-injury to assess cAMP expression levels. The corneal epithelium was treated with 1× cell lysis buffer and centrifuged at 10,000 rpm for 10 minutes to obtain the supernatant. The cAMP concentration was determined using a Cyclic AMP XP Assay Kit (Cell Signaling Technology, Danvers, MA, USA) following the manufacturer's instructions, and absorbance was measured using a microplate reader. 
Statistical Analyses
Statistical analyses were conducted using Prism 8 (GraphPad, Boston, MA, USA). Data are presented as the mean ± standard error. For normally distributed data, differences between or among groups were compared using two-tailed unpaired Student's t-test or one-way ANOVA. For non-normally distributed data, a non-parametric test (Wilcoxon rank-sum test) was used. The levels of significance are indicated as follows: ns, no significance; *P < 0.05; **P < 0.01; and ***P < 0.001 (statistical significance). All experiments were validated using at least three replicates. 
Results
NEAT1 Deficiency Contributes to Corneal Epithelial Wound Healing in Mice
We conducted a systematic analysis of lncRNA profiles in four human corneas derived from two healthy individuals using our previously published single-cell RNA-seq data to explore their biological functions in the cornea.14 Unsupervised clustering was used to annotate four primary cell types, including corneal epithelial cells, corneal stromal cells, corneal endothelial cells, and immune cells, based on classical specific markers using uniform manifold approximation and projection (UMAP) (Fig. 1A). MALAT1 and NEAT1 were the most highly expressed lncRNAs across the different subtypes and were among the most abundant genes (Fig. 1B). To further investigate the role of MALAT1 and NEAT1 in corneal epithelial wound healing, which is crucial for maintaining corneal function, we scraped the central corneal epithelium from the eyes of age-matched WT, MALAT1 knockout (M-KO),36 and NEAT1 N-KO mice (Supplementary Fig. S1). Interestingly, N-KO mice, but not M-KO mice, displayed accelerated epithelial wound closure at 12 and 24 hours post-wounding (Figs. 1C–1F). 
Figure 1.
 
NEAT1 knockout promotes corneal epithelial wound healing in mice. (A) UMAP clustering depicting the spatial distribution of human corneal cells with distinct subtypes represented by different colors. (B) Violin plot showing the expression levels of the top 10 lncRNAs for each subtype. (C, E) Corneal fluorescein staining of M-KO and N-KO mice at 0, 12, 24, and 36 hours after epithelial debridement. (D, F) Analysis of the epithelial defect area using ImageJ (n = 8).
Figure 1.
 
NEAT1 knockout promotes corneal epithelial wound healing in mice. (A) UMAP clustering depicting the spatial distribution of human corneal cells with distinct subtypes represented by different colors. (B) Violin plot showing the expression levels of the top 10 lncRNAs for each subtype. (C, E) Corneal fluorescein staining of M-KO and N-KO mice at 0, 12, 24, and 36 hours after epithelial debridement. (D, F) Analysis of the epithelial defect area using ImageJ (n = 8).
NEAT1 Knockout Promotes Corneal Nerve Regeneration and Suppresses Inflammatory Cell Infiltration
Corneal re-epithelialization is a complex, multistep process involving nerve regeneration and an inflammatory response.37,38 We investigated the role of NEAT1 in corneal nerve function and inflammation by analyzing corneal subbasal nerve fiber density and inflammatory cytokine levels. N-KO mice exhibited increased regeneration of subbasal nerve fibers compared with WT mice (Figs. 2A, 2B). Moreover, N-KO mice showed alleviated pathology after epithelial debridement, including decreased inflammatory cell infiltration (Fig. 2C) and reduced mRNA expression of CD45, IL-6, and IL-1β (Fig. 2D). These findings suggest that NEAT1 negatively regulates corneal epithelial and nerve regeneration. 
Figure 2.
 
NEAT1 deficiency improves corneal nerve regeneration and alleviates inflammation. (A) Representative images of neuronal β-tubulin Ⅲ–stained subbasal nerve fibers in corneas from WT and NEAT1 knockout mice 5 days after epithelium removal. Scale bar: 500 µm. (B) Quantification of nerve densities in the entire cornea by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 6). (C, D) H&E staining of corneal sections and mRNA expression levels of CD45, IL-6, and IL-1β in WT and NEAT1 knockout mice 24 hours after epithelial debridement (n = 3). Scale bar: 100 µm.
Figure 2.
 
NEAT1 deficiency improves corneal nerve regeneration and alleviates inflammation. (A) Representative images of neuronal β-tubulin Ⅲ–stained subbasal nerve fibers in corneas from WT and NEAT1 knockout mice 5 days after epithelium removal. Scale bar: 500 µm. (B) Quantification of nerve densities in the entire cornea by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 6). (C, D) H&E staining of corneal sections and mRNA expression levels of CD45, IL-6, and IL-1β in WT and NEAT1 knockout mice 24 hours after epithelial debridement (n = 3). Scale bar: 100 µm.
NEAT1 Knockout Activates the cAMP Signaling Pathway During Corneal Re-Epithelialization
We investigated the cellular mechanism underlying the accelerated corneal epithelial regeneration observed in NEAT1 deficiency by conducting bulk RNA-seq of the corneal epithelium from WT and N-KO mice 24 hours after epithelial debridement (Fig. 3A). RNA-seq identified 119 upregulated and 38 downregulated genes (Fig. 3B). KEGG enrichment analysis revealed significant upregulation changes in cell growth–related categories, including the RAP1, cAMP, and RAS pathways, in N-KO corneal epithelium (Fig. 3C). Subsequently, we studied the cAMP signaling pathway, which regulates corneal epithelial proliferation, migration, and apoptosis.3941 To validate this comparative analysis, we assessed the mRNA expression of cAMP pathway–related genes (Atp1a2, Ppp1r1b, Calm4, and Cngb1) and cAMP production in the corneal epithelium. As shown in Figures 3D and 3E, NEAT1 deficiency resulted in significant activation of the cAMP signaling pathway. 
Figure 3.
 
The cAMP signaling pathway is activated in NEAT1 knockout mice. (A) Flowchart illustrating the experiments conducted in this study. (B) Volcano plot depicting differentially expressed genes in the corneal epithelium of WT and NEAT1 knockout mice 24 hours after wounding (n = 4). Identification of cAMP pathway–associated mRNAs. (C) KEGG analysis depicting upregulated mRNAs. (D) qPCR validation of the upregulation of Atp1a2, Ppp1r1b, Calm4, and Cngb1 (n = 3). (E) cAMP production in the corneal epithelium (n = 3).
Figure 3.
 
The cAMP signaling pathway is activated in NEAT1 knockout mice. (A) Flowchart illustrating the experiments conducted in this study. (B) Volcano plot depicting differentially expressed genes in the corneal epithelium of WT and NEAT1 knockout mice 24 hours after wounding (n = 4). Identification of cAMP pathway–associated mRNAs. (C) KEGG analysis depicting upregulated mRNAs. (D) qPCR validation of the upregulation of Atp1a2, Ppp1r1b, Calm4, and Cngb1 (n = 3). (E) cAMP production in the corneal epithelium (n = 3).
Blockade of cAMP Signaling Inhibits Accelerated Corneal Epithelial Repair in NEAT1 Knockout Mice
To further confirm the role of cAMP signaling in promoting corneal epithelial wound healing, we used SQ22536, an effective adenylate cyclase inhibitor, to specifically disrupt cAMP signaling activation. Subconjunctival injection of 200 µm SQ22536 in N-KO mice resulted in a significant reduction in cAMP production (Supplementary Fig. S2A). Moreover, the administration of SQ22536 in N-KO mice significantly reduced the rate of epithelial repair compared with that in sham-operated animals at 12 and 24 hours post-wounding (Figs. 4A, 4B). Next, NEAT1-targeting shRNAs were injected into WT mice to specifically knockdown NEAT1 expression in the eyes before SQ22536 administration (Supplementary Figs. S2B, S3A–S3C). Consistent with the results obtained in the N-KO mice, SQ22536 delayed corneal epithelial wound healing in the N-KD mice (Figs. 4C, 4D). 
Figure 4.
 
The cAMP pathway inhibitor SQ22536 delays corneal epithelial wound healing in NEAT1 knockout or knockdown mice. (A, C) Corneal fluorescein staining of NEAT1 knockout or knockdown mice at 0, 12, and 24 hours after epithelial debridement, following subconjunctival injection of SQ22356. (B, D) Analysis of the epithelial defect area using ImageJ (N-KO, n = 5; N-KD, n = 4). NC, negative control shRNA; N-KD, NEAT1 knockdown shRNA.
Figure 4.
 
The cAMP pathway inhibitor SQ22536 delays corneal epithelial wound healing in NEAT1 knockout or knockdown mice. (A, C) Corneal fluorescein staining of NEAT1 knockout or knockdown mice at 0, 12, and 24 hours after epithelial debridement, following subconjunctival injection of SQ22356. (B, D) Analysis of the epithelial defect area using ImageJ (N-KO, n = 5; N-KD, n = 4). NC, negative control shRNA; N-KD, NEAT1 knockdown shRNA.
Activation of the cAMP Pathway Regulates Nerve Regeneration and Inflammation
Given the profound effects of NEAT1 knockout on nerve regeneration and inflammation (Fig. 2), we investigated the influence of cAMP activity on these mechanisms. Consistent with the findings in N-KO mice, N-KD mice also exhibited significantly increased subbasal nerve fiber density and weakened inflammatory responses. Compared with N-KO or N-KD mice, SQ22536-treated mice showed nerve density and gene expression similar to those of the WT or control mice (Figs. 56), suggesting that NEAT1 deficiency exerts tissue-protective effects by modulating cAMP pathway activity. 
Figure 5.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KO mice. (A) Representative images depicting wholemount-stained corneal subbasal nerve fibers in WT and NEAT1 knockout mice with or without SQ22536 5 days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve density was quantified by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Figure 5.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KO mice. (A) Representative images depicting wholemount-stained corneal subbasal nerve fibers in WT and NEAT1 knockout mice with or without SQ22536 5 days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve density was quantified by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Figure 6.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KD mice. (A) Representative images depicting wholemount-stained corneal sub-basal nerve fibers in control and NEAT1 knockdown mice with or without SQ22536, five days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve densities were quantified by analyzing the areas staining positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Figure 6.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KD mice. (A) Representative images depicting wholemount-stained corneal sub-basal nerve fibers in control and NEAT1 knockdown mice with or without SQ22536, five days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve densities were quantified by analyzing the areas staining positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Discussion
This study investigated the function of NEAT1 in the regulation of corneal re-epithelialization and nerve regeneration. Our findings indicate that NEAT1 deficiency accelerates corneal epithelial wound healing via activation of the cAMP signaling pathway. 
lncRNAs have diverse biological functions and are known to play a significant role in tissue repair and injury. MALAT1 upregulation can stimulate the inflammatory response by targeting the miR-146a/nuclear factor-κB (NF-κB) signaling pathway in lipopolysaccharide (LPS)-induced kidney injury both in vitro and in vivo.42 Furthermore, PRINS may inhibit RANTES and improve acute tubular necrosis and inflammation in renal ischemia–reperfusion injury.43 NEAT1 promotes the release of inflammatory factors and corneal neovascularization progression during corneal tissue repair.21 Moreover, NEAT1 protects the corneal endothelium from ultraviolet A (UVA)-induced FECD.14 This study provides novel evidence to support the hypothesis that NEAT1 plays a crucial role in corneal epithelial integrity and repair. 
Recent studies have highlighted the significant role of lncRNAs in neuronal regeneration. Silencing BC089918 and uc.217 using small interfering RNA (siRNA) significantly promoted the outgrowth of dorsal root ganglion neurites.44 Silc1, a conserved lncRNA, regulates nerve regeneration by cis-activating SOX11.45 In contrast, lncRNA Arrl1 inhibits axon regeneration via an lncRNA–mRNA–microRNA co-expression network.46 The deficiency of NEAT1 contributes to corneal nerve regeneration during corneal wound healing, which is mediated by activation of the cAMP signaling pathway in the corneal epithelium. However, corneal nerve fibers are primarily sensory in nature and originate from the nasociliary branch of the ophthalmic division of the trigeminal ganglion (TG). The precise role of NEAT1 in nerve regeneration and the TG remains unclear and requires further investigation. 
The cAMP pathway is a ubiquitous and versatile signaling pathway in eukaryotic cells that regulates diverse cellular functions in almost all tissues. cAMP-dependent protein kinase A (PKA) activity has been observed in corneal epithelial and endothelial cells.47 The proliferation of the corneal epithelium is endogenously regulated by the balance between adrenergic cAMP-dependent and cholinergic cGMP-dependent pathways.48 In bovine corneal endothelial cells, depolarization of plasma membrane potential activates the cAMP/PKA pathway, leading to cytoskeletal reorganization.49 The cAMP pathway regulates various cellular processes, such as the cell cycle, proliferation, differentiation, microtubule dynamics, intracellular transport, and ion fluxes.5054 This study revealed the contribution of cAMP signaling in the promotion of corneal epithelial wound healing. For the possible molecular mechanisms of cAMP overstimulation, previous studies have shown the activation of serotonin (5-HT7),55 endothelin-1,56 epidermal growth factor,57 and β-adrenergic receptors47 could activate the cAMP pathway in the cornea epithelium. Even so, the regulation mechanism upon NEAT1 deficiency requires further exploration in follow-up studies. 
Collectively, the knockout or specific knockdown of NEAT1 in the corneal epithelium of mice enhances the regeneration rates of both epithelial and nerve tissues while reducing inflammatory cell infiltration. Moreover, RNA-seq has revealed a crucial role for increased cAMP signaling in the wound repair process. These findings are potentially valuable for understanding the function of NEAT1 in corneal epithelial wound healing and the development of therapeutic strategies for corneal epithelial diseases. 
Acknowledgments
The authors especially thank Qingjun Zhou (State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Qingdao, China) for project advice and helpful discussions. 
Supported by grants from the National Natural Science Foundation of China (81800876, 82101092, 82201154). 
Disclosure: T. Sang, None; Y. Wang, None; Z. Wang, None; D. Sun, None; S. Dou, None; Y. Yu, None; X. Wang, None; C. Zhao, None; Q. Wang, None 
References
Sridhar MS. Anatomy of cornea and ocular surface. Indian J Ophthalmol. 2018; 66(2): 190–194. [CrossRef] [PubMed]
Ahmed F, House RJ, Feldman BH. Corneal abrasions and corneal foreign bodies. Prim Care. 2015; 42(3): 363–375. [CrossRef] [PubMed]
Barrientez B, Nicholas SE, Whelchel A, Sharif R, Hjortdal J, Karamichos D. Corneal injury: clinical and molecular aspects. Exp Eye Res. 2019; 186: 107709. [CrossRef] [PubMed]
Mobaraki M, Abbasi R, Vandchali SO, Ghaffari M, Moztarzadeh F, Mozafari M. Corneal repair and regeneration: current concepts and future directions. Front Bioeng Biotechnol. 2019; 7 : 135 [CrossRef] [PubMed]
Wilson SE, Liu JJ, Mohan RR. Stromal-epithelial interactions in the cornea. Prog Retin Eye Res. 1999; 18(3): 293–309. [CrossRef] [PubMed]
Ljubimov AV, Saghizadeh M. Progress in corneal wound healing. Prog Retin Eye Res. 2015; 49: 17–45. [CrossRef] [PubMed]
Suzuki K, Saito J, Yanai R, et al. Cell–matrix and cell–cell interactions during corneal epithelial wound healing. Prog Retin Eye Res. 2003; 22(2): 113–133. [CrossRef] [PubMed]
Vaidyanathan U, Hopping GC, Liu HY, et al. Persistent corneal epithelial defects: a review article. Med Hypothesis Discov Innov Ophthalmol. 2019; 8(3): 163–176. [PubMed]
Chen LL, Carmichael GG. Long noncoding RNAs in mammalian cells: what, where, and why? Wiley Interdiscip Rev RNA. 2010; 1(1): 2–21. [CrossRef] [PubMed]
Caley DP, Pink RC, Trujillano D, Carter DRF. Long noncoding RNAs, chromatin, and development. Sci World J. 2010; 10: 90–102. [CrossRef]
Ghafouri-Fard S, Abak A, Talebi SF, et al. Role of miRNA and lncRNAs in organ fibrosis and aging. Biomed Pharmacother. 2021; 143: 112132. [CrossRef] [PubMed]
Mattick JS, Taft RJ, Faulkner GJ. A global view of genomic information - moving beyond the gene and the master regulator. Trends Genet. 2010; 26(1): 21–28. [CrossRef] [PubMed]
Qureshi IA, Mattick JS, Mehler MF. Long non-coding RNAs in nervous system function and disease. Brain Res. 2010; 1338: 20–35. [CrossRef] [PubMed]
Wang Q, Dou SQ, Zhang B, et al. Heterogeneity of human corneal endothelium implicates lncRNA NEAT1 in Fuchs endothelial corneal dystrophy. Mol Ther Nucleic Acids. 2022; 27: 880–893. [CrossRef] [PubMed]
Hauser MA, Aboobakar IF, Liu YT, et al. Genetic variants and cellular stressors associated with exfoliation syndrome modulate promoter activity of a lncRNA within the LOXL1 locus. Hum Mol Genet. 2015; 24(22): 6552–6563. [CrossRef] [PubMed]
Ye W, Ma JY, Wang F, et al. LncRNA MALAT1 regulates miR-144-3p to facilitate epithelial-mesenchymal transition of lens epithelial cells via the ROS/NRF2/Notch1/Snail pathway. Oxid Med Cell Longev. 2020; 2020: 23. [CrossRef]
Yang S, Li H, Yao HP, et al. Long noncoding RNA ERLR mediates epithelial-mesenchymal transition of retinal pigment epithelial cells and promotes experimental proliferative vitreoretinopathy. Cell Death Differ. 2021; 28(8): 2351–2366. [CrossRef] [PubMed]
He XY, Chai PW, Li F, et al. A novel LncRNA transcript, RBAT1, accelerates tumorigenesis through interacting with HNRNPL and cis-activating E2F3. Mol Cancer. 2020; 19(1): 115. [CrossRef] [PubMed]
Zhong W, Yang JP, Li MY, Li L, Li AP. Long noncoding RNA NEAT1 promotes the growth of human retinoblastoma cells via regulation of miR-204/CXCR4 axis. J Cell Physiol. 2019; 234(7): 11567–11576. [CrossRef] [PubMed]
Hu XJ, Li F, He JH, et al. LncRNA NEAT1 recruits SFPQ to regulate MITF splicing and control RPE cell proliferation. Invest Ophthalmol Vis Sci. 2021; 62(14): 18. [CrossRef]
Bai YH, Lv Y, Wang WQ, Sun GL, Zhang HH. LncRNA NEAT1 promotes inflammatory response and induces corneal neovascularization. J Mol Endocrinol. 2018; 61(4): 231–239. [CrossRef] [PubMed]
Guo C, Yu M, Liu J, Jia Z, Liu H, Zhao S. Molecular mechanism of Wilms tumour 1-associated protein in diabetes-related dry eye disease by mediating m6A methylation modification of lncRNA NEAT1. J Drug Target. 2024; 32(2): 200–212. [CrossRef] [PubMed]
Ye L, Shi H, Yu C, et al. LncRNA Neat1 positively regulates MAPK signaling and is involved in the pathogenesis of Sjögren's syndrome. Int Immunopharmacol. 2020; 88: 106992. [CrossRef] [PubMed]
Satija R, Farrell JA, Gennert D, Schier AF, Regev A. Spatial reconstruction of single-cell gene expression data. Nat Biotechnol. 2015; 33(5): 495–502. [CrossRef] [PubMed]
Stuart T, Butler A, Hoffman P, et al. Comprehensive integration of single-cell data. Cell. 2019; 177(7): 1888–1902.e21. [CrossRef] [PubMed]
Yang LL, Di GH, Qi X, et al. Substance P promotes diabetic corneal epithelial wound healing through molecular mechanisms mediated via the neurokinin-1 receptor. Diabetes. 2014; 63(12): 4262–4274. [CrossRef] [PubMed]
Di GH, Du XL, Qi X, et al. Mesenchymal stem cells promote diabetic corneal epithelial wound healing through TSG-6-dependent stem cell activation and macrophage switch. Invest Ophthalmol Vis Sci. 2017; 58(10): 4344–4354. [CrossRef] [PubMed]
Zhang Z, Yang L, Li Y, et al. Interference of sympathetic overactivation restores limbal stem/progenitor cells function and accelerates corneal epithelial wound healing in diabetic mice. Biomed Pharmacother. 2023; 161: 114523. [CrossRef] [PubMed]
Wang Y, Zhao X, Wu X, Dai Y, Chen P, Xie L. microRNA-182 mediates Sirt1-induced diabetic corneal nerve regeneration. Diabetes. 2016; 65(7): 2020–2031. [CrossRef] [PubMed]
Chen S, Zhou Y, Chen Y, Gu J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics. 2018; 34(17): i884–i890. [CrossRef] [PubMed]
Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015; 12(4): 357–360. [CrossRef] [PubMed]
Roberts A, Trapnell C, Donaghey J, Rinn JL, Pachter L. Improving RNA-Seq expression estimates by correcting for fragment bias. Genome Biol. 2011; 12(3): R22. [CrossRef] [PubMed]
Anders S, Pyl PT, Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015; 31(2): 166–169. [CrossRef] [PubMed]
Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014; 15(12): 550. [CrossRef] [PubMed]
Kanehisa M, Araki M, Goto S, et al. KEGG for linking genomes to life and the environment. Nucleic Acids Res. 2008; 36: D480–D484. [CrossRef] [PubMed]
Qiao YJ, Zhang B, Wang YN, et al. Role of lncRNA MALAT1 in UVA-induced corneal endothelial senescence. Genes Dis. 2023; 10(5): 1795–1798. [CrossRef] [PubMed]
Trabucchi G, Brancato R, Verdi M, Carones F, Sala C. Corneal nerve damage and regeneration after excimer laser photokeratectomy in rabbit eyes. Invest Ophthalmol Vis Sci. 1994; 35(1): 229–235. [PubMed]
Wilson SE, Mohan RR, Mohan RR, Ambrósio R, Jr, Hong J, Lee J. The corneal wound healing response: cytokine-mediated interaction of the epithelium, stroma, and inflammatory cells. Prog Retin Eye Res. 2001; 20(5): 625–637. [CrossRef] [PubMed]
Murataeva N, Miller S, Dhopeshwarkar A, et al. Cannabinoid CB2R receptors are upregulated with corneal injury and regulate the course of corneal wound healing. Exp Eye Res. 2019; 182: 74–84. [CrossRef] [PubMed]
Nakamura M, Nishida T. Potentiation by cyclic AMP of the stimulatory effect of epidermal growth factor on corneal epithelial migration. Cornea. 2003; 22(4): 355–358. [CrossRef] [PubMed]
Wang L, Lu L. Pathway-specific effect of caffeine on protection against UV irradiation-induced apoptosis in corneal epithelial cells. Invest Ophthalmol Vis Sci. 2007; 48(2): 652–660. [CrossRef] [PubMed]
Ding Y, Guo F, Zhu T, et al. Mechanism of long non-coding RNA MALAT1 in lipopolysaccharide-induced acute kidney injury is mediated by the miR-146a/NF-κB signaling pathway. Int J Mol Med. 2018; 41(1): 446–454. [PubMed]
Yu TM, Palanisamy K, Sun KT, et al. RANTES mediates kidney ischemia reperfusion injury through a possible role of HIF-1α and LncRNA PRINS. Sci Rep. 2016; 6: 18424. [CrossRef] [PubMed]
Yao C, Wang J, Zhang HH, et al. Long non-coding RNA uc.217 regulates neurite outgrowth in dorsal root ganglion neurons following peripheral nerve injury. Eur J Neurosci. 2015; 42(1): 1718–1725. [CrossRef] [PubMed]
Perry RB, Hezroni H, Goldrich MJ, Ulitsky I. Regulation of neuroregeneration by long noncoding RNAs. Mol Cell. 2018; 72(3): 553–567. [CrossRef] [PubMed]
Wang D, Chen YP, Liu MW, et al. The long noncoding RNA Arrl1 inhibits neurite outgrowth by functioning as a competing endogenous RNA during neuronal regeneration in rats. J Biol Chem. 2020; 295(25): 8374–8386. [CrossRef] [PubMed]
Grueb M, Bartz-Schmidt KU, Rohrbach JM. Adrenergic regulation of cAMP/protein kinase A pathway in corneal epithelium and endothelium. Ophthalmic Res. 2008; 40(6): 322–328. [CrossRef] [PubMed]
Cavanagh HD, Colley AM. Cholinergic, adrenergic, and PGE1 effects on cyclic nucleotides and growth in cultured corneal epithelium. Metab Pediatr Syst Ophthalmol. 1982; 6(2): 63–74. [PubMed]
Evans F, Hernández JA, Chifflet S. Signaling pathways in cytoskeletal responses to plasma membrane depolarization in corneal endothelial cells. J Cell Physiol. 2020; 235(3): 2947–2962. [CrossRef] [PubMed]
Dumont JE, Jauniaux JC, Roger PP. The cyclic AMP-mediated stimulation of cell proliferation. Trends Biochem Sci. 1989; 14(2): 67–71. [CrossRef] [PubMed]
Bertolotto C, Abbe P, Hemesath TJ, et al. Microphthalmia gene product as a signal transducer in cAMP-induced differentiation of melanocytes. J Cell Biol. 1998; 142(3): 827–835. [CrossRef] [PubMed]
Sehrawat S, Ernandez T, Cullere X, et al. AKAP9 regulation of microtubule dynamics promotes Epac1-induced endothelial barrier properties. Blood. 2011; 117(2): 708–718. [CrossRef] [PubMed]
Pathak A, Del Monte F, Zhao W, et al. Enhancement of cardiac function and suppression of heart failure progression by inhibition of protein phosphatase 1. Circ Res. 2005; 96(7): 756–766. [CrossRef] [PubMed]
Guba M, Kuhn M, Forssmann WG, Classen M, Gregor M, Seidler U. Guanylin strongly stimulates rat duodenal HCO3 secretion: proposed mechanism and comparison with other secretagogues. Gastroenterology. 1996; 111(6): 1558–1568. [CrossRef] [PubMed]
Grueb M, Rohrbach JM, Schlote T, Mielke J. Serotonin (5-HT7) receptor-stimulated activation of cAMP-PKA pathway in bovine corneal epithelial and endothelial cells. Ophthalmic Res. 2012; 48(1): 22–27. [CrossRef] [PubMed]
Socci RR, Tachado SD, Aronstam RS, Reinach PS. Characterization of the muscarinic receptor subtypes in the bovine corneal epithelial cells. J Ocul Pharmacol Ther. 1996; 12(3): 259–269. [CrossRef] [PubMed]
Yang H, Wang Z, Miyamoto Y, Reinach PS. Cell signaling pathways mediating epidermal growth factor stimulation of Na:K:2Cl cotransport activity in rabbit corneal epithelial cells. J Membr Biol. 2001; 182(2): 93–101. [CrossRef]
Figure 1.
 
NEAT1 knockout promotes corneal epithelial wound healing in mice. (A) UMAP clustering depicting the spatial distribution of human corneal cells with distinct subtypes represented by different colors. (B) Violin plot showing the expression levels of the top 10 lncRNAs for each subtype. (C, E) Corneal fluorescein staining of M-KO and N-KO mice at 0, 12, 24, and 36 hours after epithelial debridement. (D, F) Analysis of the epithelial defect area using ImageJ (n = 8).
Figure 1.
 
NEAT1 knockout promotes corneal epithelial wound healing in mice. (A) UMAP clustering depicting the spatial distribution of human corneal cells with distinct subtypes represented by different colors. (B) Violin plot showing the expression levels of the top 10 lncRNAs for each subtype. (C, E) Corneal fluorescein staining of M-KO and N-KO mice at 0, 12, 24, and 36 hours after epithelial debridement. (D, F) Analysis of the epithelial defect area using ImageJ (n = 8).
Figure 2.
 
NEAT1 deficiency improves corneal nerve regeneration and alleviates inflammation. (A) Representative images of neuronal β-tubulin Ⅲ–stained subbasal nerve fibers in corneas from WT and NEAT1 knockout mice 5 days after epithelium removal. Scale bar: 500 µm. (B) Quantification of nerve densities in the entire cornea by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 6). (C, D) H&E staining of corneal sections and mRNA expression levels of CD45, IL-6, and IL-1β in WT and NEAT1 knockout mice 24 hours after epithelial debridement (n = 3). Scale bar: 100 µm.
Figure 2.
 
NEAT1 deficiency improves corneal nerve regeneration and alleviates inflammation. (A) Representative images of neuronal β-tubulin Ⅲ–stained subbasal nerve fibers in corneas from WT and NEAT1 knockout mice 5 days after epithelium removal. Scale bar: 500 µm. (B) Quantification of nerve densities in the entire cornea by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 6). (C, D) H&E staining of corneal sections and mRNA expression levels of CD45, IL-6, and IL-1β in WT and NEAT1 knockout mice 24 hours after epithelial debridement (n = 3). Scale bar: 100 µm.
Figure 3.
 
The cAMP signaling pathway is activated in NEAT1 knockout mice. (A) Flowchart illustrating the experiments conducted in this study. (B) Volcano plot depicting differentially expressed genes in the corneal epithelium of WT and NEAT1 knockout mice 24 hours after wounding (n = 4). Identification of cAMP pathway–associated mRNAs. (C) KEGG analysis depicting upregulated mRNAs. (D) qPCR validation of the upregulation of Atp1a2, Ppp1r1b, Calm4, and Cngb1 (n = 3). (E) cAMP production in the corneal epithelium (n = 3).
Figure 3.
 
The cAMP signaling pathway is activated in NEAT1 knockout mice. (A) Flowchart illustrating the experiments conducted in this study. (B) Volcano plot depicting differentially expressed genes in the corneal epithelium of WT and NEAT1 knockout mice 24 hours after wounding (n = 4). Identification of cAMP pathway–associated mRNAs. (C) KEGG analysis depicting upregulated mRNAs. (D) qPCR validation of the upregulation of Atp1a2, Ppp1r1b, Calm4, and Cngb1 (n = 3). (E) cAMP production in the corneal epithelium (n = 3).
Figure 4.
 
The cAMP pathway inhibitor SQ22536 delays corneal epithelial wound healing in NEAT1 knockout or knockdown mice. (A, C) Corneal fluorescein staining of NEAT1 knockout or knockdown mice at 0, 12, and 24 hours after epithelial debridement, following subconjunctival injection of SQ22356. (B, D) Analysis of the epithelial defect area using ImageJ (N-KO, n = 5; N-KD, n = 4). NC, negative control shRNA; N-KD, NEAT1 knockdown shRNA.
Figure 4.
 
The cAMP pathway inhibitor SQ22536 delays corneal epithelial wound healing in NEAT1 knockout or knockdown mice. (A, C) Corneal fluorescein staining of NEAT1 knockout or knockdown mice at 0, 12, and 24 hours after epithelial debridement, following subconjunctival injection of SQ22356. (B, D) Analysis of the epithelial defect area using ImageJ (N-KO, n = 5; N-KD, n = 4). NC, negative control shRNA; N-KD, NEAT1 knockdown shRNA.
Figure 5.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KO mice. (A) Representative images depicting wholemount-stained corneal subbasal nerve fibers in WT and NEAT1 knockout mice with or without SQ22536 5 days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve density was quantified by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Figure 5.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KO mice. (A) Representative images depicting wholemount-stained corneal subbasal nerve fibers in WT and NEAT1 knockout mice with or without SQ22536 5 days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve density was quantified by analyzing the areas that stained positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Figure 6.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KD mice. (A) Representative images depicting wholemount-stained corneal sub-basal nerve fibers in control and NEAT1 knockdown mice with or without SQ22536, five days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve densities were quantified by analyzing the areas staining positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Figure 6.
 
SQ22536 reverses the improvement in corneal nerve regeneration and inflammation in N-KD mice. (A) Representative images depicting wholemount-stained corneal sub-basal nerve fibers in control and NEAT1 knockdown mice with or without SQ22536, five days after epithelium removal. Scale bar: 500 µm. (B) Corneal nerve densities were quantified by analyzing the areas staining positive for β-tubulin Ⅲ (n = 4). (C) Validation of IL-6 and IL-1β expression levels using qPCR (n = 3).
Table 1.
 
Target Sequences
Table 1.
 
Target Sequences
Table 2.
 
Primer Pairs Used for qRT-PCR Analyses
Table 2.
 
Primer Pairs Used for qRT-PCR Analyses
×
×

This PDF is available to Subscribers Only

Sign in or purchase a subscription to access this content. ×

You must be signed into an individual account to use this feature.

×