Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Exp Eye Res ; 242: 109862, 2024 May.
Article in English | MEDLINE | ID: mdl-38490292

ABSTRACT

The continual exposure of retinal tissues to oxidative stress leads to discernible anatomical and physiological alterations. Specifically, the onslaught of oxidative damage escalates the irreversible death of retinal pigmented epithelium (RPE) cells, pinpointed as the fundamental pathological event in dry age-related macular degeneration (AMD). There is a conspicuous lack of effective therapeutic strategies to counteract this degenerative process. This study screened a library of antioxidants for their ability to protect RPE cells against oxidative stress and identified L-ergothioneine (EGT) as a potent cytoprotective agent. L-ergothioneine provided efficient protection against oxidative stress-damaged RPE and maintained cell redox homeostasis and normal physiological functions. It maintained the normal structure of the retina in mice under oxidative stress conditions. Transcriptomic analysis revealed that EGT counteracted major gene expression changes induced by oxidative stress. It upregulated antioxidant gene expression and inhibited NRF2 translocation. The inhibition of NRF2 abolished EGT's protective effects, suggesting that NRF2 activation contributes to its mechanism of action. In conclusion, we identified EGT as a safe and effective small-molecule compound that is expected to be a novel antioxidative agent for treating AMD.


Subject(s)
Antioxidants , Ergothioneine , NF-E2-Related Factor 2 , Oxidative Stress , Retinal Pigment Epithelium , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Animals , Ergothioneine/pharmacology , Antioxidants/pharmacology , Oxidative Stress/drug effects , Mice , Mice, Inbred C57BL , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology , Cells, Cultured , Humans , Blotting, Western , Disease Models, Animal , Gene Expression Regulation/drug effects , Reactive Oxygen Species/metabolism
2.
Aging Cell ; : e14247, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38887148

ABSTRACT

The corneal epithelium is the outermost transparent barrier of the eyeball and undergoes continuous self-renewal by limbal stem cells (LSCs) during its lifetime; however, the impact of aging on LSCs remains largely unknown. Here, we showed that the healing ability of the cornea in elderly macaques (Macaca fascicularis) was significantly decreased compared to that of younger macaques. This delayed wound closure accompanied a disordered cell arrangement and corneal opacity. A novel cytokine, Secreted and Transmembrane 1 (SECTM1), was found to facilitate corneal healing and was upregulated in young macaques upon wounding. Mechanistically, SECTM1 is essential for LSC migration and proliferation, and may partially function through Cell Division Cycle Associated 7 (CDCA7). Notably, the topical application of SECTM1 to aged wounded corneas dramatically promoted re-epithelialization and improved corneal transparency in both mice and macaques. Our work suggests that aging may impair the expression of healing response factors and injury repair in non-human primate corneas, and that SECTM1 application could potentially benefit corneal wound healing in clinical treatment.

3.
Invest Ophthalmol Vis Sci ; 65(6): 17, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38861275

ABSTRACT

Purpose: N6-methyladenosine (m6A) methylation is a chemical modification that occurs on RNA molecules, where the hydrogen atom of adenine (A) nucleotides is replaced by a methyl group, forming N6-methyladenosine. This modification is a dynamic and reversible process that plays a crucial role in regulating various biological processes, including RNA stability, transport, translation, and degradation. Currently, there is a lack of research on the role of m6A modifications in maintaining the characteristics of RPE cells. m6A readers play a crucial role in executing the functions of m6A modifications, which prompted our investigation into their regulatory roles in the RPE. Methods: Phagocytosis assays, immunofluorescence staining, flow cytometry experiments, ß-galactosidase staining, and RNA sequencing (RNA-seq) were conducted to assess the functional and cellular characteristics changes in retinal pigment epithelium (RPE) cells following short-hairpin RNA-mediated knockdown of insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2). RNA-seq and ultraviolet crosslinking immunoprecipitation with high-throughput sequencing (HITS-CLIP) were employed to identify the target genes regulated by IGF2BP2. adeno-associated virus (AAV) subretinal injection was performed in 6- to 8-week-old C57 mice to reduce IGF2BP2 expression in the RPE, and the impact of IGF2BP2 knockdown on mouse visual function was assessed using immunofluorescence, quantitative real-time PCR, optical coherence tomography, and electroretinography. Results: IGF2BP2 was found to have a pronounced effect on RPE phagocytosis. Subsequent in-depth exploration revealed that IGF2BP2 modulates the mRNA stability of PAX6 and OTX2, and the loss of IGF2BP2 induces inflammatory and aging phenotypes in RPE cells. IGF2BP2 knockdown impaired RPE function, leading to retinal dysfunction in vivo. Conclusions: Our data suggest a crucial role of IGF2BP2 as an m6A reader in maintaining RPE homeostasis by regulating the stability of PAX6 and OTX2, making it a potential target for preventing the occurrence of retinal diseases related to RPE malfunction.


Subject(s)
Otx Transcription Factors , PAX6 Transcription Factor , RNA-Binding Proteins , Retinal Pigment Epithelium , Animals , Mice , Cells, Cultured , Electroretinography , Flow Cytometry , Gene Expression Regulation/physiology , Homeostasis , Mice, Inbred C57BL , Otx Transcription Factors/metabolism , Otx Transcription Factors/genetics , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Phagocytosis/physiology , Retinal Pigment Epithelium/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Tomography, Optical Coherence
4.
Nat Commun ; 15(1): 256, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38177186

ABSTRACT

Proper differentiation of corneal epithelial cells (CECs) from limbal stem/progenitor cells (LSCs) is required for maintenance of ocular homeostasis and clear vision. Here, using a single-cell transcriptomic atlas, we delineate the comprehensive and refined molecular regulatory dynamics during human CEC development and differentiation. We find that RORA is a CEC-specific molecular switch that initiates and drives LSCs to differentiate into mature CECs by activating PITX1. RORA dictates CEC differentiation by establishing CEC-specific enhancers and chromatin interactions between CEC gene promoters and distal regulatory elements. Conversely, RORA silences LSC-specific promoters and disrupts promoter-anchored chromatin loops to turn off LSC genes. Collectively, our work provides detailed and comprehensive insights into the transcriptional dynamics and RORA-mediated epigenetic remodeling underlying human corneal epithelial differentiation.


Subject(s)
Cornea , Epigenomics , Humans , Cell Differentiation/genetics , Gene Expression Profiling , Chromatin/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1
5.
Cell Prolif ; 56(9): e13433, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36851859

ABSTRACT

Limbal stem/progenitor cells (LSC) represent the source of corneal epithelium renewal. LSC proliferation and differentiation are essential for corneal homeostasis, however, the regulatory mechanism remains largely unexplored. Here, we performed single-cell RNA sequencing and discovered proliferation heterogeneity as well as spontaneously differentiated and senescent cell subgroups in multiply passaged primary LSC. Fasciculation and elongation protein zeta 1 (FEZ1) and Dickkopf-1 (DKK1) were identified as two significant regulators of LSC proliferation and senescence. These two factors were mainly expressed in undifferentiated corneal epithelial cells (CECs). Knocking down the expression of either FEZ1 or DKK1 reduced cell division and caused cell cycle arrest. We observed that DKK1 acted as a downstream target of FEZ1 in LSC and that exogenous DKK1 protein partially prevented growth arrest and senescence upon FEZ1 suppression in vitro. In a mouse model of corneal injury, DKK1 also rescued the corneal epithelium after recovery was inhibited by FEZ1 suppression. Hence, the FEZ1-DKK1 axis was required for CEC proliferation and the juvenile state and can potentially be targeted as a therapeutic strategy for promoting recovery after corneal injury.


Subject(s)
Adaptor Proteins, Signal Transducing , Corneal Injuries , Intercellular Signaling Peptides and Proteins , Limbal Stem Cells , Nerve Tissue Proteins , Transcriptome , Animals , Mice , Cell Proliferation , Corneal Injuries/metabolism , Limbal Stem Cells/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism
6.
Adv Mater ; 35(16): e2207750, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36680510

ABSTRACT

Corneal transplantation is impeded by donor shortages, immune rejection, and ethical reservations. Pre-made cornea prostheses (keratoprostheses) offer a proven option to alleviate these issues. Ideal keratoprostheses must possess optical clarity and mechanical robustness, but also high permeability, processability, and recyclability. Here, it is shown that rationally controlling the extent of arrested phase separation can lead to optimized multiscale structure that reconciles permeability and transparency, a previously conflicting goal by common pore-forming strategies. The process is simply accomplished by hydrothermally treating a dense and transparent hydrophobic association hydrogel. The examination of multiscale structure evolution during hydrothermal treatment reveals that the phase separation with upper miscibility gap evolves to confer time-dependent pore growth due to slow dynamics of polymer-rich phase which is close to vitrification. Such a process can render a combination of multiple desired properties that equal or surpass those of the state-of-the-art keratoprostheses. In vivo tests confirm that the keratoprosthesis can effectively repair corneal perforation and restore a transparent cornea with treatment outcomes akin to that of allo-keratoplasty. The keratoprosthesis is easy to access and convenient to carry, and thus would be an effective temporary substitute for a corneal allograft in emergency conditions.


Subject(s)
Cornea , Corneal Diseases , Humans , Cornea/surgery , Prostheses and Implants , Corneal Diseases/surgery , Hydrogels/chemistry , Treatment Outcome
7.
Transl Vis Sci Technol ; 11(6): 28, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35771535

ABSTRACT

Purpose: Wound healing of the corneal epithelium mainly involves two types of cells: limbal stem/progenitor cells (LSCs) and differentiated central corneal epithelial cells (CECs). The healing ability of CECs is still debatable, and its correlated transcriptomic alterations during wound healing are yet to be elucidated. This study aimed to determine the healing ability and mechanisms underlying the actions of CECs using rabbit ocular surface injury models. Methods: A central corneal ring-like residual epithelium model was used to investigate the healing ability of CECs. Uninjured and injury-stimulated LSCs and CECs were collected for transcriptomic analysis. The analysis results were verified by quantitative reverse transcriptase polymerase chain reaction, immunofluorescence staining, and two types of rabbit corneal injury models. Results: During wound healing, the upregulated genes in LSCs were mostly enriched in the mitotic cell cycle-related processes, but those in CECs were mostly enriched in cell adhesion and migration. CECs could repair the epithelial defects successfully at one-time injuries. However, after repetitive injuries, the CECs repaired notably slower and failed to completely heal the defect, but the LSCs repaired even faster than the one-time injury. Conclusions: Our results indicated rabbit CECs repair the epithelial defect mainly depending on migration and its proliferative ability is limited, and LSCs are the main source of regenerative epithelial cells. Translational Relevance: This study provides information on gene expression in the corneal epithelium during wound healing, indicating that regulation of the cell cycle, cell adhesion, and migration may be the basis for future treatment strategies for corneal wound healing.


Subject(s)
Corneal Injuries , Epithelium, Corneal , Animals , Cell Differentiation , Cornea , Corneal Injuries/metabolism , Epithelium, Corneal/metabolism , Rabbits , Stem Cells/metabolism
8.
Invest Ophthalmol Vis Sci ; 63(2): 14, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35129588

ABSTRACT

Purpose: Cornea, the outermost transparent layer of the eye, is the first line of defense against external threats. Following injury, the wound healing response is crucial to corneal repair and regeneration, yet its underlying mechanism is poorly understood. Our study was designed to investigate the role of dsRNA and its regulatory network in corneal wound healing. Methods: A corneal wound healing model was established via the surgical removal of half of the corneal surface and adjoining limbus. RNase III was then used to clarify the role of dsRNA in corneal wound closure and RNA-seq was performed to investigate the mechanism of dsRNA in the healing process. Related gene expression was assessed using immunofluorescence staining, qPCR, and Western blot. Flow cytometry and scratch assay were used to analyze the proliferation and migration of limbal stem/progenitor cells (LSCs) in vitro and functional analysis of the target genes was completed using the corneal wound healing model. Results: Corneal wound healing was delayed and impaired when the dsRNAs were removed or damaged following RNase III digestion. The dsRNAs released following corneal damage activate type I interferon (IFN-I) signaling, primarily IFNß, via the corneal epithelium and neutralizing IFNß or blocking IFN-I signaling delays corneal wound closure. Moreover, our data identified MMP13 as a downstream effector of IFNß where its expression promotes LSC proliferation and enhances corneal epithelial reconstruction in vivo. Conclusions: The dsRNA induced IFNß-MMP13 axis plays a key role in corneal wound healing.


Subject(s)
Corneal Injuries/genetics , Epithelium, Corneal/pathology , Interleukin-6/genetics , Matrix Metalloproteinase 13/genetics , Mutation , RNA-Binding Proteins/genetics , RNA/genetics , Wound Healing/genetics , Animals , Cells, Cultured , Corneal Injuries/diagnosis , Corneal Injuries/metabolism , DNA Mutational Analysis , Disease Models, Animal , Epithelium, Corneal/injuries , Epithelium, Corneal/metabolism , Interleukin-6/metabolism , Matrix Metalloproteinase 13/metabolism , Mice , RNA-Binding Proteins/metabolism
9.
Ophthalmol Sci ; 2(2): 100148, 2022 Jun.
Article in English | MEDLINE | ID: mdl-36249679

ABSTRACT

Purpose: To propose an improved stem cell-based strategy for limbal stem cell deficiency (LSCD) treatment. Design: Experimental randomized or parallel-group animal study. Subjects: Fifty adult male New Zealand white rabbits. Methods: Human limbal stem/progenitor cells (LSCs) and limbal stromal stem/progenitor cells (LSSCs) were cultured in serum-free conditions and further differentiated into corneal epithelial cells and keratocytes, respectively. All cell types were characterized with lineage-specific markers. Gene expression analysis was performed to identify the potential function of LSSCs in corneal regeneration. Two LSCD models of rabbits for transplantations were used: transplantation performed at the time of limbal and corneal epithelial excision (LSCD model) and transplantation performed after clinical signs were induced in an LSCD model (pLSCD model). The pLSCD model better mimics the pathologic changes and symptoms of human LSCD. Rabbit models received LSC or LSC plus LSSC treatment. Corneal epithelial defects, neovascularization, and opacity were assessed every 3 weeks for 24 weeks. ZsGreen-labeled LSSCs were used for short-term tracking in vivo. Main Outcome Measures: Rates of corneal epithelial defect area, corneal neovascularization and opacity scores, graft survival rate, and immunofluorescence staining of specific markers. Results: Both LSC transplantation and LSC plus LSSC cotransplantation effectively repaired the corneal surface in the LSCD model. These 2 strategies showed no significant differences in terms of graft survival rate or epithelial repair. However, corneal opacity was observed in the LSC group (in 3 of 8 rabbits), but not in the LSC plus LSSC group. Notably, when treating LSCD rabbits with distinguishable stromal opacification and neovascularization, cotransplantation of LSCs and LSSCs exhibited significantly better therapeutic effects than transplantation of LSCs alone, with graft survival rates of 87.5% and 37.5%, respectively. The implanted LSSCs could differentiate into keratocytes during the wound-healing process. RNA sequencing analysis showed that the stromal cells produced not only a collagen-rich extracellular matrix to facilitate reconstruction of the lamellar structure, but also niche factors that accelerated epithelial cell growth and inhibited angiogenesis and inflammation. Conclusions: These findings highlight the support of stromal cells in niche homeostasis and tissue regeneration, providing LSC plus LSSC cotransplantation as a new treatment strategy for corneal blindness.

10.
Sci Adv ; 8(28): eabo5668, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35857527

ABSTRACT

Understanding the regulatory network of cell fate acquisition remains a major challenge. Using the induction of surface epithelium (SE) from human embryonic stem cells as a paradigm, we show that the dynamic changes in morphology-related genes (MRGs) closely correspond to SE fate transitions. The marked remodeling of cytoskeleton indicates the initiation of SE differentiation. By integrating promoter interactions, epigenomic features, and transcriptome, we delineate an SE-specific cis-regulatory network and identify grainyhead-like 3 (GRHL3) as an initiation factor sufficient to drive SE commitment. Mechanically, GRHL3 primes the SE chromatin accessibility landscape and activates SE-initiating gene expression. In addition, the evaluation of GRHL3-mediated promoter interactions unveils a positive feedback loop of GRHL3 and bone morphogenetic protein 4 on SE fate decisions. Our work proposes a concept that MRGs could be used to identify cell fate transitions and provides insights into regulatory principles of SE lineage development and stem cell-based regenerative medicine.

11.
Signal Transduct Target Ther ; 5(1): 20, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32296021

ABSTRACT

Accumulated oxidative damage may lead to irreversible retinal pigmented epithelium (RPE) cell death, which is considered to be the primary cause of dry age-related macular degeneration (AMD), leading to blindness in the elderly. However, an effective therapy for this disease is lacking. Here, we described a robust high-content screening procedure with a library of 814 protective compounds and found that D609 strongly protected RPE cells from sodium iodate (SI)-induced oxidative cell death and prolonged their healthy survival. D609 effectively attenuated excessive reactive oxygen species (ROS) and prevented severe mitochondrial loss due to oxidative stress in the RPE cells. Surprisingly, the potent antioxidative effects of D609 were not achieved through its own reducibility but were primarily dependent on its ability to increase the expression of metallothionein. The injection of this small water-soluble molecule also showed an explicit protective effect of the RPE layer in an SI-induced AMD mouse model. These findings suggested that D609 could serve as a novel antioxidative protector of RPE cells both in vitro and in vivo and unveiled a novel antioxidative mechanism of D609, which may ultimately have clinical applications for the treatment of AMD.


Subject(s)
Macular Degeneration/drug therapy , Mitochondria/drug effects , Oxidative Stress/drug effects , Retinal Pigment Epithelium/drug effects , Animals , Cell Death/drug effects , Disease Models, Animal , Humans , Macular Degeneration/genetics , Macular Degeneration/pathology , Mitochondria/genetics , Norbornanes/pharmacology , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/pathology , Thiocarbamates/pharmacology
13.
Nat Genet ; 45(12): 1504-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24162740

ABSTRACT

Vitamin C, a micronutrient known for its anti-scurvy activity in humans, promotes the generation of induced pluripotent stem cells (iPSCs) through the activity of histone demethylating dioxygenases. TET hydroxylases are also dioxygenases implicated in active DNA demethylation. Here we report that TET1 either positively or negatively regulates somatic cell reprogramming depending on the absence or presence of vitamin C. TET1 deficiency enhances reprogramming, and its overexpression impairs reprogramming in the context of vitamin C by modulating the obligatory mesenchymal-to-epithelial transition (MET). In the absence of vitamin C, TET1 promotes somatic cell reprogramming independent of MET. Consistently, TET1 regulates 5-hydroxymethylcytosine (5hmC) formation at loci critical for MET in a vitamin C-dependent fashion. Our findings suggest that vitamin C has a vital role in determining the biological outcome of TET1 function at the cellular level. Given its benefit to human health, vitamin C should be investigated further for its role in epigenetic regulation.


Subject(s)
Ascorbic Acid/pharmacology , Cellular Reprogramming/drug effects , DNA-Binding Proteins/physiology , Proto-Oncogene Proteins/physiology , Animals , Cells, Cultured , Embryo, Mammalian , Epigenesis, Genetic/drug effects , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout
14.
Cell Res ; 21(6): 884-94, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21445094

ABSTRACT

The ectopic expression of several transcription factors can restore embryonic cell fate to cultured somatic cells and generate induced pluripotent stem cells (iPSCs), revealing a previously unknown pathway to pluripotency. However, this technology is currently limited by low efficiency, slow kinetics and multi-factorial requirement. Here we show that reprogramming can be improved and dramatically accelerated by optimizing culture conditions. First, we developed an optimized defined medium, iCD1, which allows Oct4/Sox2/Klf4 (OSK)-mediated reprogramming to achieve ultra-high efficiency (~10% at day 8). We also found that this optimized condition renders both Sox2 and Klf4 dispensable, although the elimination of these two factors leads to lower efficiency and slower kinetics. Our studies define a shortened route, both in timing and factor requirement, toward pluripotency. This new paradigm not only provides a rationale to further improve iPSC generation but also simplifies the conceptual understanding of reprogramming by defined factors.


Subject(s)
Cell Dedifferentiation/genetics , Culture Media/chemistry , Induced Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Proliferation , Coculture Techniques , Culture Media/metabolism , Fibroblasts/metabolism , Genetic Vectors , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Karyotyping , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred ICR , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retroviridae/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Time Factors , Transfection , Transplantation Chimera
SELECTION OF CITATIONS
SEARCH DETAIL