Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Am J Physiol Cell Physiol ; 326(5): C1367-C1383, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38406826

ABSTRACT

Age-related macular degeneration (AMD) is characterized by the degenerative senescence in the retinal pigment epithelium (RPE) and photoreceptors, which is accompanied by the accumulation of iron ions in the aging retina. However, current models of acute oxidative stress are still insufficient to simulate the gradual progression of AMD. To address this, we established chronic injury models by exposing the aRPE-19 cells, 661W cells, and mouse retina to iron ion overload over time. Investigations at the levels of cell biology and molecular biology were performed. It was demonstrated that long-term treatment of excessive iron ions induced senescence-like morphological changes, decreased cell proliferation, and impaired mitochondrial function, contributing to apoptosis. Activation of the mitogen-activated protein kinase (MAPK) pathway and the downstream molecules were confirmed both in the aRPE-19 and 661W cells. Furthermore, iron ion overload resulted in dry AMD-like lesions and decreased visual function in the mouse retina. These findings suggest that chronic exposure to overloading iron ions plays a significant role in the pathogenesis of retinopathy and provide a potential model for future studies on AMD.NEW & NOTEWORTHY To explore the possibility of constructing reliable research carriers on age-related macular degeneration (AMD), iron ion overload was applied to establish models in vitro and in vivo. Subsequent investigations into cellular physiology and molecular biology confirmed the presence of senescence in these models. Through this study, we hope to provide a better option of feasible methods for future researches into AMD.


Subject(s)
Disease Models, Animal , Iron , Macular Degeneration , Retinal Pigment Epithelium , Animals , Humans , Macular Degeneration/metabolism , Macular Degeneration/pathology , Macular Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Mice , Iron/metabolism , Mice, Inbred C57BL , Apoptosis , Oxidative Stress , Cell Line , Cellular Senescence , Iron Overload/metabolism , Iron Overload/pathology , Cell Proliferation , Retina/metabolism , Retina/pathology , Mitochondria/metabolism , Mitochondria/pathology
2.
Mol Biol Rep ; 51(1): 477, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38573426

ABSTRACT

BACKGROUND: The retinal pigment epithelium (RPE) is essential for retinal homeostasis. Comprehensively exploring the transcriptional patterns of diabetic human RPE promotes the understanding of diabetic retinopathy (DR). METHODS AND RESULTS: A total of 4125 differentially expressed genes (DEGs) were screened out from the human primary RPE cells subjected to prolonged high glucose (HG). The subsequent bioinformatics analysis is divided into 3 steps. In Step 1, 21 genes were revealed by intersecting the enriched genes from the KEGG, WIKI, and Reactome databases. In Step 2, WGCNA was applied and intersected with the DEGs. Further intersection based on the enrichments with the GO biological processes, GO cellular components, and GO molecular functions databases screened out 12 candidate genes. In Step 3, 13 genes were found to be simultaneously up-regulated in the DEGs and a GEO dataset involving human diabetic retinal tissues. VEGFA and ERN1 were the 2 starred genes finally screened out by overlapping the 3 Steps. CONCLUSION: In this study, multiple genes were identified as crucial in the pathological process of RPE under protracted HG, providing potential candidates for future researches on DR. The current study highlights the importance of RPE in DR pathogenesis.


Subject(s)
Diabetic Retinopathy , Retina , Humans , Diabetic Retinopathy/genetics , Epithelial Cells , Retinal Pigments , Glucose
3.
Hum Genet ; 142(1): 103-123, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36129575

ABSTRACT

Mutations in myelin regulatory factor (MYRF), a gene mapped to 11q12-q13.3, are responsible for autosomal dominant high hyperopia and seem to be associated with angle closure glaucoma, which is one of the leading causes of irreversible blindness worldwide. Whether there is a causal link from the MYRF mutations to the pathogenesis of primary angle-closure glaucoma (PACG) remains unclear at this time. Six truncation mutations, including five novel and one previously reported, in MYRF are identified in seven new probands with hyperopia, of whom all six adults have glaucoma, further confirming the association of MYRF mutations with PACG. Immunofluorescence microscopy demonstrates enriched expression of MYRF in the ciliary body and ganglion cell layer in humans and mice. Myrfmut/+ mice have elevated IOP and fewer ganglion cells along with thinner retinal nerve fiber layer with ganglion cell layer than wild-type. Transcriptome sequencing of Myrfmut/+ retinas shows downregulation of Dnmt3a, a gene previously associated with PACG. Co-immunoprecipitation demonstrates a physical association of DNMT3A with MYRF. DNA methylation sequencing identifies several glaucoma-related cell events in Myrfmut/+ retinas. The interaction between MYRF and DNMT3A underlies MYRF-associated PACG and provides clues for pursuing further investigation into the pathogenesis of PACG and therapeutic target.


Subject(s)
Eye Diseases, Hereditary , Glaucoma, Angle-Closure , Hyperopia , Humans , Adult , Mice , Animals , Hyperopia/genetics , Glaucoma, Angle-Closure/genetics , Glaucoma, Angle-Closure/complications , Mutation , Eye Diseases, Hereditary/genetics , Transcription Factors/genetics , Intraocular Pressure/genetics
4.
Exp Eye Res ; 233: 109524, 2023 08.
Article in English | MEDLINE | ID: mdl-37290629

ABSTRACT

Mitogen-activated protein kinase kinase kinase kinase-4 (MAP4K4) is a potential regulator of photoreceptor development. To investigate the mechanisms underlying MAP4K4 during the neuronal development of retinal photoreceptors, we generated knockout models of C57BL/6j mice in vivo and 661 W cells in vitro. Our findings revealed homozygous lethality and neural tube malformation in mice subjected to Map4k4 DNA ablation, providing evidence for the involvement of MAP4K4 in early stage embryonic neural formation. Furthermore, our study demonstrated that the ablation of Map4k4 DNA led to the vulnerability of photoreceptor neurites during induced neuronal development. By monitoring transcriptional and protein variations in mitogen-activated protein kinase (MAPK) signaling pathway-related factors, we discovered an imbalance in neurogenesis-related factors in Map4k4 -/- cells. Specifically, MAP4K4 promotes jun proto-oncogene (c-JUN) phosphorylation and recruits other factors related to nerve growth, ultimately leading to the robust formation of photoreceptor neurites. These data suggest that MAP4K4 plays a decisive role in regulating the fate of retinal photoreceptors through molecular modulation and contributes to our understanding of vision formation.


Subject(s)
Neurogenesis , Signal Transduction , Animals , Mice , DNA , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate , NF-kappaB-Inducing Kinase
5.
FASEB J ; 36(10): e22577, 2022 10.
Article in English | MEDLINE | ID: mdl-36165267

ABSTRACT

Oxidative stress-induced damage to and dysfunction of retinal pigment epithelium (RPE) cells are important pathogenetic factors of age-related macular degeneration (AMD); however, the underlying molecular mechanism is not fully understood. Long noncoding RNAs (lncRNAs) have important roles in various biological processes. In this study, using an oxidative damage model in RPE cells, we identified a novel oxidation-related lncRNA named CYLD-AS1. We further revealed that the expression of CYLD-AS1 was increased in RPEs during oxidative stress. Depletion of CYLD-AS1 promoted cell proliferation and mitochondrial function and protected RPE cells against hydrogen peroxide (H2 O2 )-induced damage. Mechanistically, CYLD-AS1 also regulated the expression of NRF2, which is related to oxidative stress, and NF-κB signaling pathway members, which are related to inflammation. Remarkably, these two signaling pathways were mediated by the CYLD-AS1 interactor miR-134-5p. Moreover, exosomes secreted by CYLD-AS1 knockdown RPE cells had a lower proinflammatory effect than those secreted by control cells. In summary, our study revealed that CYLD-AS1 affects the oxidative stress-related and inflammatory functions of RPE cells by sponging miR-134-5p to mediate NRF2/NF-κB signaling pathway activity, suggesting that targeting CYLD-AS1 could be a promising strategy for the treatment of AMD and related diseases.


Subject(s)
Macular Degeneration , MicroRNAs , RNA, Long Noncoding , Deubiquitinating Enzyme CYLD/genetics , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Inflammation/metabolism , Macular Degeneration/metabolism , MicroRNAs/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oxidative Stress , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Retinal Pigment Epithelium/metabolism , Signal Transduction/genetics
6.
Int J Mol Sci ; 23(19)2022 Oct 09.
Article in English | MEDLINE | ID: mdl-36233291

ABSTRACT

IL-24 is a multifunctional cytokine that regulates both immune cells and epithelial cells. Although its elevation is associated with a number of autoimmune diseases, its tolerogenic properties against autoreactive T cells have recently been revealed in an animal model of central nervous system (CNS) autoimmunity by inhibiting the pathogenic Th17 response. To explore the potential of IL-24 as a therapeutic agent in CNS autoimmunity, we induced experimental autoimmune uveitis (EAU) in wildtype mice and intravitreally injected IL-24 into the inflamed eye after disease onset. We found that the progression of ocular inflammation was significantly inhibited in the IL-24-treated eye when compared to the control eye. More importantly, IL-24 treatment suppressed cytokine production from ocular-infiltrating, pathogenic Th1 and Th17 cells. In vitro experiments confirmed that IL-24 suppressed both Th1 and Th17 differentiation by regulating their master transcription factors T-bet and RORγt, respectively. In addition, we found that intravitreal injection of IL-24 suppressed the production of proinflammatory cytokines and chemokines from the retinas of the EAU-inflamed eyes. This observation appears to be applicable in humans, as IL-24 similarly inhibits human retinal pigment epithelium cells ARPE-19. In conclusion, we report here that IL-24, as a multifunctional cytokine, is capable of resolving ocular inflammation in EAU mice by targeting both uveitogenic T cells and RPE cells. This study sheds new light on IL-24 as a potential therapeutic candidate for autoimmune uveitis.


Subject(s)
Autoimmune Diseases , Uveitis , Animals , Autoimmunity , Cytokines/therapeutic use , Disease Models, Animal , Humans , Inflammation/pathology , Interleukins , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3 , Retina/pathology , Th1 Cells , Th17 Cells , Uveitis/pathology
7.
J Cell Mol Med ; 25(9): 4220-4234, 2021 05.
Article in English | MEDLINE | ID: mdl-33759344

ABSTRACT

Proliferative vitreoretinopathy (PVR) is a refractory vitreoretinal fibrosis disease, and epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) cells is the key pathological mechanism of PVR. However, few studies focused on the role of METTL3, the dominating methyltransferase for m6A RNA modification in PVR pathogenesis. Immunofluorescence staining and qRT-PCR were used to determine the expression of METTL3 in human tissues. Lentiviral transfection was used to stably overexpress and knockdown METTL3 in ARPE-19 cells. MTT assay was employed to study the effects of METTL3 on cell proliferation. The impact of METTL3 on the EMT of ARPE-19 cells was assessed by migratory assay, morphological observation and expression of EMT markers. Intravitreal injection of cells overexpressing METTL3 was used to assess the impact of METTL3 on the establishment of the PVR model. We found that METTL3 expression was less in human PVR membranes than in the normal RPE layers. In ARPE-19 cells, total m6A abundance and the METTL3 expression were down-regulated after EMT. Additionally, METTL3 overexpression inhibited cell proliferation through inducing cell cycle arrest at G0/G1 phase. Furthermore, METTL3 overexpression weakened the capacity of TGFß1 to trigger EMT by regulating wnt/ß -catenin pathway. Oppositely, knockdown of METTL3 facilitated proliferation and EMT of ARPE-19 cells. In vivo, intravitreal injection of METTL3-overexpressing cells delayed the development of PVR compared with injection of control cells. In summary, this study suggested that METTL3 is involved in the PVR process, and METTL3 overexpression inhibits the EMT of ARPE-19 cells in vitro and suppresses the PVR process in vivo.


Subject(s)
Epithelial-Mesenchymal Transition , Methyltransferases/metabolism , Retinal Pigment Epithelium/pathology , Vitreoretinopathy, Proliferative/prevention & control , Wnt Proteins/metabolism , beta Catenin/metabolism , Adolescent , Adult , Aged , Female , Follow-Up Studies , Gene Expression Regulation , Humans , Male , Methyltransferases/genetics , Middle Aged , Prognosis , Retinal Pigment Epithelium/metabolism , Vitreoretinopathy, Proliferative/metabolism , Vitreoretinopathy, Proliferative/pathology , Wnt Proteins/genetics , Young Adult , beta Catenin/genetics
8.
BMC Ophthalmol ; 21(1): 235, 2021 May 27.
Article in English | MEDLINE | ID: mdl-34044792

ABSTRACT

PURPOSE AND BACKGROUND: Recently, we found that maximal medial rectus recession and lateral rectus resection in patients with complete lateral rectus paralysis resulted in a partial restoration of abduction. In an attempt to understand some of the mechanisms involved with this effect we examined gene expression profiles of lateral recti from these patients, with our focus being directed to genes related to myogenesis. MATERIALS AND METHODS: Lateral recti resected from patients with complete lateral rectus paralysis and those from concomitant esotropia (controls) were collected. Differences in gene expression profiles between these two groups were examined using microarray analysis and quantitative Reverse-transcription PCR (qRT-PCR). RESULTS: A total of 3056 differentially expressed genes (DEGs) were identified between these two groups. Within the paralytic esotropia group, 2081 genes were up-regulated and 975 down-regulated. The results of RT-PCR revealed that PAX7, MYOG, PITX1, SIX1 and SIX4 showed higher levels of expression, while that of MYOD a lower level of expression within the paralytic esotropia group as compared with that in the control group (p < 0.05). CONCLUSION: The decreased expression of MYOD in the paralytic esotropia group suggested that extraocular muscle satellite cell (EOMSCs) differentiation processes were inhibited. Whereas the high expression levels of PAX7, SIX1/4 and MYOG, suggested that the EOMSCs were showing an effective potential for differentiation. The stimulation resulting from muscle surgery may induce EOMSCs to differentiate and thus restore abduction function.


Subject(s)
Abducens Nerve Diseases , Esotropia , Cell Differentiation , Esotropia/surgery , Homeodomain Proteins , Humans , Oculomotor Muscles/surgery , Ophthalmologic Surgical Procedures , Retrospective Studies
9.
Ophthalmic Res ; 64(2): 337-344, 2021.
Article in English | MEDLINE | ID: mdl-32344402

ABSTRACT

INTRODUCTION: Paralytic strabismus involves a functional loss of extraocular muscles resulting from muscular or neuronal disorders. Currently, only a limited number of drugs are available for functional repair of extraocular muscles. Here, we investigated the effects of a novel drug, flavonoids sophoranone, on the differentiation of extraocular muscles as assessed in bothin vivo and in vitro models. MATERIALS AND METHODS: The effect of flavonoids sophoranone on C2C12 cells was examinedin vitro as evaluated with use of apoptosis, reactive oxygen species (ROS), and cell viability assays. Then, both in vivo and in vitro effects of this drug were examined on the differentiation of C2C12 and satellite cells within extraocular muscles in rabbits. For these latter experiments, RT-PCR and Western blot assays were used to determine expression levels of markers for myogenic differentiation. RESULTS: With use of flavonoids sophoranone concentrations ranging from 0 to 10 µM, no effects were observed upon cell apoptosis, ROS, and cell cycle in C2C12 cells. Based on MTT assay results, flavonoids sophoranone was shown to increase C2C12 cell proliferation. Moreover, flavonoids sophoranone promoted the differentiation of C2C12 and satellite cells within extraocular muscles in rabbits, which were verified as based on cell morphology and expression levels of mRNA and protein markers of myogenic differentiation. Finally, flavonoids sophoranone treatment also increased gene expressions of Myh3, Myog, and MCK. CONCLUSION: The capacity for flavonoids sophoranone to upgrade the differentiation of both C2C12 and satellite cells within extraocular muscles in rabbits at concentrations producing no adverse effects suggest that this drug may provide a safe and effective means to promote repair of damaged extraocular muscles.


Subject(s)
Apoptosis , Flavonoids/pharmacology , Muscle Development/genetics , Myoblasts/drug effects , Oculomotor Muscles/cytology , Animals , Cell Cycle , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Models, Animal , Myoblasts/cytology , Myoblasts/metabolism , Oculomotor Muscles/drug effects , Oculomotor Muscles/metabolism , Rabbits , Reactive Oxygen Species/metabolism
10.
J Cell Mol Med ; 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33090698

ABSTRACT

Retinoblastoma (RB) is a common intraocular malignancy in children. Due to the poor prognosis of RB, it is crucial to search for efficient diagnostic and therapeutic strategies. Studies have shown that methyltransferase-like 3 (METTL3), a major RNA N (6)-adenosine methyltransferase, is closely related to the initiation and development of cancers. Nevertheless, whether METTL3 is associated with RB remains unexplored. Therefore, we investigated the function and mechanisms of METTL3 in the regulation of RB progression. We manipulated METTL3 expression in RB cells. Then, cell proliferation, apoptosis, migration and invasion were analysed. We also analysed the expression of PI3K/AKT/mTOR pathway members. Finally, we incorporated subcutaneous xenograft mouse models into our studies. The results showed that METTL3 is highly expressed in RB patients and RB cells. We found that METTL3 knockdown decreases cell proliferation, migration and invasion of RB cells, while METTL3 overexpression promotes RB progression in vitro and in vivo. Moreover, two downstream members of the PI3K/AKT/mTOR pathway, P70S6K and 4EBP1, were affected by METTL3. Our study revealed that METTL3 promotes the progression of RB through PI3K/AKT/mTOR pathways in vitro and in vivo. Targeting the METTL3/PI3K/AKT/mTOR signalling axis could be a promising therapeutic strategy for the treatment of RB.

11.
Arch Biochem Biophys ; 650: 1-13, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29753724

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of blindness during aging. The degeneration of retinal pigment epithelium (RPE) is the main pathologic characteristic of AMD. ID2 is a member of the Inhibitor of DNA binding proteins (ID) family and is involved in regulation of cell proliferation and differentiation. However, currently the role of ID2 in oxidative injury response in RPE cells remains unknown. Here we showed that oxidative stress increased ID2 expression in RPE cells. Knockdown of ID2 promoted cell apoptosis and increased ROS level in RPE cells that were subjected to oxidative damage. In addition, over-expression of ID2 attenuated the oxidative damage response in RPE cells. Mechanistically, ID2 protected RPE cells from oxidative damage through activating NRF2. Furthermore, phosphorylation of ERK1/2 positively regulated the protective function of ID2. Finally, we confirmed that the oxidative damage increased Id2 expression and over-expression of Id2 elevated Nrf2 expression in primary mouse RPE cells. Therefore, ID2 protects RPE cells from oxidative damage through the p-ERK1/2/ID2/NRF2 pathway. Our study contributes to a better understanding of the mechanisms underlying oxidative stress in AMD and may present a new strategy for AMD treatment.


Subject(s)
Inhibitor of Differentiation Protein 2/metabolism , MAP Kinase Signaling System , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Retinal Pigment Epithelium/metabolism , Signal Transduction , Animals , Apoptosis , Cell Line , Cells, Cultured , Gene Knockdown Techniques , Humans , Inhibitor of Differentiation Protein 2/genetics , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice, Inbred C57BL , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/pathology , Up-Regulation
12.
Mol Vis ; 23: 579-587, 2017.
Article in English | MEDLINE | ID: mdl-28848320

ABSTRACT

PURPOSE: To observe the nuclear expression and interaction of heparanase and RNA polymerase II (RNA Pol II), an enzyme that catalyzes the transcription of DNA in eukaryotic cells) in human retinal microvascular endothelial cells (HRECs) under high glucose condition and to investigate the association of heparanase with the transcription activity of the vascular endothelial growth factor (VEGF) gene promoter. METHODS: Cultured HRECs were maintained for 3 days in media with high or normal glucose. The expressions of heparanase and RNA Pol II in each group were analyzed with immunofluorescence. Co-immunoprecipitation was applied to detect the interaction of heparanase and Pol II proteins. Cells in both groups were used for chromatin immunoprecipitation (ChIP) with anti-heparanase and anti-RNA Pol II antibodies to identify high-confidence heparanase-binding regions across the entire VEGF gene promoter. Moreover, real-time PCR was used to demonstrate the interaction between heparanase and the VEGF gene promoter region. RESULTS: The immunofluorescence studies showed that the nuclear expression of heparanase was intense in high-glucose HRECs but faint in the normal group; RNA Pol II in the nucleus was also intense in high glucose HRECs, and the distribution of heparanase was consistent with that of RNA Pol II. The co-immunoprecipitation data showed that heparanase combined with RNA Pol II in HRECs cells treated with high glucose, and the molecular size of HPA interacted with RNA Pol II was 50 kDa, while no combination of two proteins was evident in normal HRECs cells. Real-time PCR-based ChIP results showed that the high-confidence HPA-binding region was -1155 to -1018 (containing hypoxia response element) in the VEGF gene promoter, and the cells treated with high glucose showed increases in heparanase and RNA Pol II in the VEGF gene promoter region compared with the normal glucose treated cells (t = -3.244, p = 0.032; t = -6.096, p = 0.004, respectively). CONCLUSIONS: Nuclear heparanase combines directly with the VEGF gene promoter and is involved in the regulation of VEGF gene transcription in high-glucose HRECs.


Subject(s)
Endothelial Cells/drug effects , Gene Expression Regulation/physiology , Glucose/pharmacology , Glucuronidase/metabolism , Vascular Endothelial Growth Factor A/genetics , Blotting, Western , Cell Proliferation , Cells, Cultured , Chromatin/metabolism , Endothelial Cells/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Immunoprecipitation , RNA Polymerase II/metabolism , Real-Time Polymerase Chain Reaction , Retinal Vessels/cytology , Transcription, Genetic
13.
J Biol Chem ; 288(9): 6238-47, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23349461

ABSTRACT

SFMBT1 belongs to the malignant brain tumor domain-containing chromatin reader family that recognizes repressive histone marks and represses transcription. The biological functions and molecular basis underlying SFMBT1-mediated transcriptional repression are poorly elucidated. Here, our proteomic analysis revealed that SFMBT1 is associated with multiple transcriptional corepressor complexes, including CtBP/LSD1/HDAC complexes, polycomb repressive complexes, and malignant brain tumor family proteins, that collectively contribute to SFMBT1 repressor activity. During myogenesis, Sfmbt1 represses myogenic differentiation of cultured and primary myoblasts. Mechanistically, Sfmbt1 interacts with MyoD and mediates epigenetic silencing of MyoD target genes via recruitment of its associated corepressors and subsequent induction of epigenetic modifications and chromatin compaction. Therefore, our study identified novel mechanisms accounting for SFMBT1-mediated transcription repression and revealed an essential role of Sfmbt1 in regulating MyoD-mediated transcriptional silencing that is required for the maintenance of undifferentiated states of myogenic progenitor cells.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Gene Silencing/physiology , Muscle Development/physiology , Repressor Proteins/metabolism , Transcription, Genetic/physiology , Cell Line , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , MyoD Protein/genetics , MyoD Protein/metabolism , Proteomics/methods , Repressor Proteins/genetics
14.
J Biol Chem ; 288(38): 27680-27691, 2013 Sep 20.
Article in English | MEDLINE | ID: mdl-23928305

ABSTRACT

Chromatin readers decipher the functional readouts of histone modifications by recruiting specific effector complexes for subsequent epigenetic reprogramming. The LSD1 (also known as KDM1A) histone demethylase complex modifies chromatin and represses transcription in part by catalyzing demethylation of dimethylated histone H3 lysine 4 (H3K4me2), a mark for active transcription. However, none of its currently known subunits recognizes methylated histones. The Snai1 family transcription factors are central drivers of epithelial-to-mesenchymal transition (EMT) by which epithelial cells acquire enhanced invasiveness. Snai1-mediated transcriptional repression of epithelial genes depends on its recruitment of the LSD1 complex and ensuing demethylation of H3K4me2 at its target genes. Through biochemical purification, we identified the MBT domain-containing protein SFMBT1 as a novel component of the LSD1 complex associated with Snai1. Unlike other mammalian MBT domain proteins characterized to date that selectively recognize mono- and dimethylated lysines, SFMBT1 binds di- and trimethyl H3K4, both of which are enriched at active promoters. We show that SFMBT1 is essential for Snai1-dependent recruitment of LSD1 to chromatin, demethylation of H3K4me2, transcriptional repression of epithelial markers, and induction of EMT by TGFß. Carcinogenic metal nickel is a widespread environmental and occupational pollutant. Nickel alters gene expression and induces EMT. We demonstrate the nickel-initiated effects are dependent on LSD1-SFMBT1-mediated chromatin modification. Furthermore, in human cancer, expression of SFMBT1 is associated with mesenchymal markers and unfavorable prognosis. These results highlight a critical role of SFMBT1 in epigenetic regulation, EMT, and cancer.


Subject(s)
Chromatin/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Histone Demethylases/metabolism , Histones/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Repressor Proteins/metabolism , Carcinogens/pharmacology , Chromatin/genetics , Chromatin/pathology , Epithelial Cells/pathology , HEK293 Cells , Histone Demethylases/genetics , Histones/genetics , Humans , Methylation , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Nickel/adverse effects , Nickel/pharmacology , Repressor Proteins/genetics , Snail Family Transcription Factors , Trace Elements , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Stem Cells ; 31(4): 823-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23307608

ABSTRACT

Muscular dystrophies are a group of devastating diseases characterized by progressive muscle weakness and degeneration, with etiologies including muscle gene mutations and regenerative defects of muscle stem cells. Notch signaling is critical for skeletal myogenesis and has important roles in maintaining the muscle stem cell pool and preventing premature muscle differentiation. To investigate the functional impact of Notch signaling blockade in muscle stem cells, we developed a conditional knock-in mouse model in which endogenous Notch signaling is specifically blocked in muscle stem cell compartment. Mice with Notch signaling inhibition in muscle stem cells showed several muscular dystrophic features and impaired muscle regeneration. Analyses of satellite cells and isolated primary myoblasts revealed that Notch signaling blockade in muscle stem cells caused reduced activation and proliferation of satellite cells but enhanced differentiation of myoblasts. Our data thus indicate that Notch signaling controls processes that are critical to regeneration in muscular dystrophy, suggesting that Notch inhibitor therapies could have potential side effects on muscle functions.


Subject(s)
Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Development/physiology , Muscular Dystrophies/metabolism , Receptors, Notch/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Mice , Mice, Knockout , Muscle Development/genetics , Muscular Dystrophies/genetics , Myoblasts/cytology , Myoblasts/metabolism , Receptors, Notch/genetics , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
16.
Mol Biol Rep ; 40(1): 217-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23079703

ABSTRACT

The Six1 homeoprotein belongs to the Six (sine oculis) transcription factor family, the members of which are known to act as master regulators of development. Six1 is essential for promoting myogenesis during mammalian somitogenesis. Previous studies have shown that Six1 participates in later steps of myogenic differentiation by enhancing early activation of myogenin via binding to the Mef3 site of the myogenin promoter. In the present study, however, we show that overexpression of Six1 via retroviral infection suppresses the expression of myogenin and myosin in C2C12 myoblasts, consequently retarding myogenic differentiation without affecting cell proliferation or expression of Mef2 and Mef3. These findings further demonstrate the functional role of Six1 in myogenesis.


Subject(s)
Cell Differentiation , Homeodomain Proteins/genetics , Muscle Development/genetics , Myoblasts/cytology , Myoblasts/metabolism , Animals , Cell Line , Cell Proliferation , Gene Expression , Myogenin/genetics , Myogenin/metabolism , Myosins/genetics , Myosins/metabolism
17.
ScientificWorldJournal ; 2013: 178708, 2013.
Article in English | MEDLINE | ID: mdl-24453806

ABSTRACT

The Notch signaling is an evolutionarily conserved cell-cell communication pathway that plays critical roles in the proliferation, survival, apoptosis, and fate determination of mammalian cells. Retinal pigment epithelial (RPE) cells are responsible for supporting the function of the neural retina and maintaining vision. This study investigated the function of Notch signaling in RPE cells. We found that the members of the Notch signaling pathway components were differentially expressed in RPE cells. Furthermore, blockage of Notch signaling inhibited the migration and proliferation of RPE cells and reduced the expression levels of certain Notch signaling target genes, including HES1, MYC, HEY2, and SOX9. Our data reveal a critical role of Notch signaling in RPE cells, suggesting that targeting Notch signaling may provide a novel approach for the treatment of ophthalmic diseases related to RPE cells.


Subject(s)
Cell Movement , Cell Proliferation , Receptor, Notch1/physiology , Retinal Pigment Epithelium/cytology , Signal Transduction , Apoptosis , Cell Line , Cell Lineage , HEK293 Cells , Humans , Lentivirus/metabolism , Neurons/metabolism , Plasmids/metabolism , RNA/metabolism , Receptor, Notch1/metabolism , Retina/metabolism , Vision, Ocular
18.
Front Genet ; 14: 1231415, 2023.
Article in English | MEDLINE | ID: mdl-37867602

ABSTRACT

Stomach Adenocarcinoma (STAD) is a leading cause of death worldwide. Somatic Copy Number Alterations (SCNAs), which result in Homologous recombination (HR) deficiency in double-strand break repair, are associated with the progression of STAD. However, the landscape of frequent breakpoints of SCNAs (hotspots) and their functional impacts remain poorly understood. In this study, we aimed to explore the frequency and impact of these hotspots in 332 STAD patients and 1,043 cancer cells using data from the Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE). We studied the rates of DSB (Double-Strand Breaks) loci in STAD patients by employing the Non-Homogeneous Poisson Distribution (λ), based on which we identified 145 DSB-hotspots with genes affected. We further verified DNA cytosine deamination as a critical process underlying the burden of DSB in STAD. Finally, we illustrated the clinical impact of the significant biological processes. Our findings highlighted the relationship between DNA cytosine deamination and SCNA in cancer was associated with recurrent Somatic Copy Number Alterations in STAD.

19.
BMC Complement Med Ther ; 23(1): 55, 2023 Feb 18.
Article in English | MEDLINE | ID: mdl-36800952

ABSTRACT

BACKGROUND: Age-related macular degeneration (AMD) is a leading cause of vision loss in elderly people, and dry AMD is the most common type of AMD. Oxidative stress and alternative complement pathway activation may play essential roles in the pathogenesis of dry AMD. There are no available drugs for dry AMD. Qihuang Granule (QHG) is an herbal formula for the treatment of dry AMD, and it achieves a good clinical effect in our hospital. However, its potential mechanism is unclear. Our study investigated the effects of QHG on oxidative stress-associated retinal damage to reveal its underlying mechanism. METHODS: Oxidative stress models were established using H2O2 and NaIO3 in ARPE-19 cells and C57BL/6 mice. Cell apoptosis and viability were assessed using phase contrast microscopy and flow cytometry, respectively. Alterations in the mouse retinal structure were evaluated using Masson staining and transmission electron microscopy (TEM). The expression of complement factor H (CFH), complement component 3a (C3a) and complement component 5a (C5a) in retinal pigment epithelium (RPE) cells and mice was measured using RT‒PCR, Western blot analysis and ELISA. RESULTS: Pretreatment with QHG significantly prevented cell apoptosis and disorder of the RPE and inner segment/outer segment (IS/OS) in H2O2-treated RPE cells and NaIO3-injected mice. QHG alleviated mitochondrial damage in mouse RPE cells, as shown by TEM. QHG also promoted CFH expression and inhibited the expression of C3a and C5a. CONCLUSIONS: The results suggest that QHG protects the retinal pigment epithelium from oxidative stress, likely by regulating the alternative complement pathway.


Subject(s)
Macular Degeneration , Retinal Pigment Epithelium , Animals , Mice , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Complement Pathway, Alternative , Hydrogen Peroxide/pharmacology , Mice, Inbred C57BL , Oxidative Stress , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Macular Degeneration/pathology
20.
Invest Ophthalmol Vis Sci ; 64(13): 47, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37906058

ABSTRACT

Purpose: The purpose of this study was to investigate the effects of silibinin on epithelial-mesenchymal transition (EMT) of retinal pigment epithelial (RPE) and proliferative vitreoretinopathy (PVR) formation, as well as its underlying molecular mechanism. Methods: Cellular morphological change and EMT molecular markers were evaluated by using phase contrast imaging, qPCR, and Western blot (WB) to investigate the impact of silibinin on the EMT of ARPE-19 cells. Scratch assay and transwell assay were used to study the effect of silibinin on cell migration. An intravitreally injected RPE-induced rat PVR model was used to assess the effect of silibinin on PVR in vivo. RNA-seq was applied to study the molecular mechanism of silibinin-mediated PVR prevention. Results: Silibinin inhibited TGFß1-induced EMT and migration of RPE in a dose-dependent manner in vitro. Moreover, silibinin prevented proliferative membrane formation in an intravitreal injected RPE-induced rat PVR model. In line with these findings, RNA-seq revealed a global suppression of TGFß1-induced EMT and migration-related genes by silibinin in RPEs. Mechanistically, silibinin reduced TGFß1-induced phosphorylation levels of Smad3 and Stat3, and Smad3 nuclear translocation in RPE. Conclusions: Silibinin inhibits the EMT of RPE cells in vitro and prevents the formation of PVR membranes in vivo. Mechanistically, silibinin inhibits Smad3 phosphorylation and suppresses Smad3 nuclear translocation through the inhibition of Stat3 phosphorylation. These findings suggest that silibinin may serve as a potential treatment for PVR.


Subject(s)
Transforming Growth Factor beta , Vitreoretinopathy, Proliferative , Animals , Rats , Phosphorylation , Epithelial-Mesenchymal Transition , Vitreoretinopathy, Proliferative/drug therapy , Silybin
SELECTION OF CITATIONS
SEARCH DETAIL