Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 54
1.
Front Plant Sci ; 14: 1285616, 2023.
Article En | MEDLINE | ID: mdl-38034556

Introduction: Ainaxiang (Blumea balsamifera (Linn.) DC.) is cultivated for the extraction of (-)-borneol and other pharmaceutical raw materials due to its abundant volatile oil. However, there is limited knowledge regarding the structural basis and composition of volatile oil accumulation in fresh B. balsamifera leaves. Methods: To address this problem, we compare the fresh leaves' morphology, microstructure, and volatile metabonomic at different development stages, orderly defined from the recently unfolded young stage (S1) to the senescent stage (S4). Results and discussion: Distinct differences were observed in the macro-appearance and microstructure at each stage, particularly in the B. balsamifera glandular trichomes (BbGTs) distribution. This specialized structure may be responsible for the accumulation of volatile matter. 213 metabolites were identified through metabolomic analysis, which exhibited spatiotemporal accumulation patterns among different stages. Notably, (-)-borneol was enriched at S1, while 10 key odor metabolites associated with the characteristic balsamic, borneol, fresh, and camphor aromas of B. balsamifera were enriched in S1 and S2. Ultra-microstructural examination revealed the involvement of chloroplasts, mitochondria, endoplasmic reticulum, and vacuoles in the synthesizing, transporting, and storing essential oils. These findings confirm that BbGTs serve as the secretory structures in B. balsamifera, with the population and morphology of BbGTs potentially serving as biomarkers for (-)-borneol accumulation. Overall, young B. balsamifera leaves with dense BbGTs represent a rich (-)-borneol source, while mesophyll cells contribute to volatile oil accumulation. These findings reveal the essential oil accumulation characteristics in B. balsamifera, providing a foundation for further understanding.

2.
Talanta ; 260: 124627, 2023 Aug 01.
Article En | MEDLINE | ID: mdl-37182291

Due to the worldwide ecological and environmental issues induced by heavy metal pollution, including zinc and manganese, the ratio-metric discrimination of Zn2+ and Mn2+ based on CDs is urgently required. In this work, reduced CDs (re-CDs) with the intrinsic dual emissive peaks are obtained, and specific discrimination of Zn2+ and Mn2+ is realized by re-CDs with ratio-metric mode. With the addition of Zn2+, the fluorescent (FL) intensity at 650 nm increases obviously, while that at 680 nm progressively decreases. However, the presence of Mn2+ would induce the quenching of FL intensity at 680 nm while that at 650 nm remains constant. Then the Zn2+ and Mn2+ can be separately determined with the ratio of FL intensity at 650 nm to that at 680 mm (F650/F680). Under optimal conditions, the limit of detection (LOD) of Zn2+ is determined to be 9.09 nmol/L, and that for Mn2+ is estimated to be 0.93 nmol/L, which is much lower than previously reported work and standard level of Zn2+ and Mn2+ permitted in drinking water by WHO. Moreover, the specific recognition of Mn2+ and Zn2+ can be realized via the addition of different masking agents (ethylenediamine for Zn2+ and triethanolamine for Mn2+). Furthermore, our results reveal that the structural changes from -NH-CO to -NC-OH induced by Zn2+ contribute to the shift of FL peak from 680 to 650 nm while both static and dynamic quenching processes are involved in the detection of Mn2+. The ratio-metric probe was successfully applied to Zn2+ and Mn2+ determination in human serum samples and Sandy Lake water.

3.
CNS Neurosci Ther ; 29 Suppl 1: 146-160, 2023 06.
Article En | MEDLINE | ID: mdl-36924268

INTRODUCTION: Optic nerve injury is a leading cause of irreversible blindness worldwide. The retinal ganglion cells (RGCs) and their axons cannot be regenerated once damaged. Therefore, reducing RGC damage is crucial to prevent blindness. Accordingly, we aimed to investigate the potential influence of the gut microbiota on RGC survival, as well as the associated action mechanisms. METHODS: We evaluated the effects of microbiota, specifically Bifidobacterium, on RGC. Optic nerve crush (ONC) was used as a model of optic nerve injury. Vancomycin and Bifidobacterium were orally administered to specific pathogen-free (SPF) mice. RESULTS: Bifidobacterium promoted RGC survival and optic nerve regeneration. The administration of Bifidobacterium inhibited microglia activation but promoted Müller cell activation, which was accompanied by the downregulation of inflammatory cytokines and upregulation of neurotrophic factors and retinal ERK/Fos signaling pathway activation. CONCLUSIONS: Our study demonstrates that Bifidobacterium-induced changes in intestinal flora promote RGC survival. The protective effect of Bifidobacterium on RGC can be attributed to the inhibition of microglia activation and promotion of Müller cell activation and the secondary regulation of inflammatory and neurotrophic factors.


Optic Nerve Injuries , Retinal Ganglion Cells , Mice , Animals , Retinal Ganglion Cells/metabolism , Optic Nerve Injuries/therapy , Optic Nerve Injuries/metabolism , Neuroglia/metabolism , Axons/metabolism , Nerve Growth Factors/metabolism , Blindness/metabolism , Cell Survival/physiology , Disease Models, Animal
4.
J Bone Miner Res ; 38(4): 556-567, 2023 04.
Article En | MEDLINE | ID: mdl-36722289

Temporomandibular joint (TMJ) growth requires orchestrated interactions between various cell types. Recent studies revealed that fibrocartilage stem cells (FCSCs) in the TMJ cartilage play critical roles as cell resources for joint development and repair. However, the detailed molecular network that influences FCSC fate during TMJ cartilage development remains to be elucidated. Here, we investigate the functional role of Igf1 in FCSCs for TMJ cartilage growth and homeostasis by lineage tracing using Gli1-CreER+ ; Tmflfl mice and conditional Igf1 deletion using Gli1-/Col2-CreER+ ; Igf1fl/fl mice. In Gli1-CreER+ ; Tmflfl mice, red fluorescence+ (RFP+ ) FCSCs show a favorable proliferative capacity. Igf1 deletion in Gli1+ /Col2+ cell lineages leads to distinct pathological changes in TMJ cartilage. More serious cartilage thickness and cell density reductions are found in the superficial layers in Gli1-CreER+ ; Igf1fl/fl mice. After long-term Igf1 deletion, a severe disordered cell arrangement is found in both groups. When Igf1 is conditionally deleted in vivo, the red fluorescent protein-labeled Gli1+ FCSC shows a significant disruption of chondrogenic differentiation, cell proliferation, and apoptosis leading to TMJ cartilage disarrangement and subchondral bone loss. Immunostaining shows that pAkt signaling is blocked in all cartilage layers after the Gli1+ -specific deletion of Igf1. In vitro, Igf1 deletion disrupts FCSC capacities, including proliferation and chondrogenesis. Moreover, the deletion of Igf1 in FCSCs significantly aggravates the joint osteoarthritis phenotype in the unilateral anterior crossbite mouse model, characterized by decreased cartilage thickness and cell numbers as well as a loss of extracellular matrix secretions. These findings uncover Igf1 as a regulator of TMJ cartilage growth and repair. The deletion of Igf1 disrupts the progenitor capacity of FCSCs, leading to a disordered cell distribution and exaggerating TMJ cartilage dysfunction. © 2023 American Society for Bone and Mineral Research (ASBMR).


Cartilage, Articular , Insulin-Like Growth Factor I , Temporomandibular Joint , Animals , Mice , Cartilage, Articular/metabolism , Chondrocytes/metabolism , Fibrocartilage , Homeostasis , Stem Cells , Temporomandibular Joint/metabolism , Zinc Finger Protein GLI1/metabolism , Insulin-Like Growth Factor I/metabolism
5.
Biochem Genet ; 61(1): 187-201, 2023 Feb.
Article En | MEDLINE | ID: mdl-35817886

Atherosclerosis (AS) is a chronic inflammatory disease with high morbidity and mortality rates worldwide. This study aimed to investigate the role of circular RNA protein tyrosine phosphatase receptor type A (circRNA_PTPRA) in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cell (HUVECs) injury and its underlying molecular mechanism. The expression of circRNA-PTPRA and microRNA (miR)-671-5p was assessed by quantitative reverse transcription PCR (qRT-PCR). The interaction between circRNA-PTPRA and miR-671-5p was predicted using bioinformatic analysis. Cell viability and apoptosis were determined using the Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. Inflammation in HUVECs was analyzed by measuring the secretion of tumor necrosis factor alpha (TNF-α), interleukin-1beta (IL-1ß), and IL-6 using enzyme-linked immunosorbent assay (ELISA). Cleaved-caspase-3 expression was assessed using western blotting. The results indicated that circRNA-PTPRA expression was significantly increased and miR-671-5p expression was decreased in the serum of patients with AS and in ox-LDL-treated HUVECs. The interaction between circRNA-PTPRA and miR-671-5p was verified by dual luciferase reporter and RNA pull-down assays. In HUVECs, downregulation of circRNA-PTPRA reversed ox-LDL-induced reduction in cell viability, increase in apoptosis, and enhanced inflammation, whereas all these effects mediated by circRNA-PTPRA downregulation in ox-LDL-treated HUVECs were abolished by miR-671-5p downregulation. In conclusion, circRNA-PTPRA downregulation protects against ox-LDL-induced HUVECs injury by upregulating miR-671-5p, thereby providing potential therapeutic targets for AS.


Atherosclerosis , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, LDL/pharmacology , Apoptosis , Inflammation/genetics , Inflammation/metabolism , Atherosclerosis/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/pharmacology
6.
Sci Transl Med ; 14(647): eabj2177, 2022 06.
Article En | MEDLINE | ID: mdl-35648811

Antiangiogenesis therapies targeting vascular endothelial growth factor (VEGF) have revolutionized the treatment of neovascular ocular diseases, including neovascular age-related macular degeneration (nAMD). Compelling evidence has implicated the vital role of complement system dysregulation in AMD pathogenesis, implying it as a potential therapeutic strategy for geographic atrophy in dry AMD and to enhance the efficacy of anti-VEGF monotherapies in nAMD. This study reports the preclinical assessment and phase 1 clinical outcomes of a bispecific fusion protein, efdamrofusp alfa (code: IBI302), which is capable of neutralizing both VEGF isoforms and C3b/C4b. Efdamrofusp alfa showed superior efficacy over anti-VEGF monotherapy in a mouse laser-induced choroidal neovascularization (CNV) model after intravitreal delivery. Dual inhibition of VEGF and the complement activation was found to further inhibit macrophage infiltration and M2 macrophage polarization. Intravitreal efdamrofusp alfa demonstrated favorable safety profiles and exhibited antiangiogenetic efficacy in a nonhuman primate laser-induced CNV model. A phase 1 dose-escalating clinical trial (NCT03814291) was thus conducted on the basis of the preclinical data. Preliminary results showed that efdamrofusp alfa was well tolerated in patients with nAMD. These data suggest that efdamrofusp alfa might be effective for treating nAMD and possibly other complement-related ocular conditions.


Choroidal Neovascularization , Macular Degeneration , Angiogenesis Inhibitors/therapeutic use , Animals , Choroidal Neovascularization/drug therapy , Complement System Proteins , Eye , Humans , Macular Degeneration/complications , Macular Degeneration/drug therapy , Mice , Vascular Endothelial Growth Factor A/therapeutic use
7.
Exp Mol Med ; 54(5): 673-684, 2022 05.
Article En | MEDLINE | ID: mdl-35624154

Subretinal fibrosis remains a major obstacle to the management of neovascular age-related macular degeneration. Choroidal pericytes were found to be a significant source of subretinal fibrosis, but the underlying mechanisms of pericyte-myofibroblast transition (PMT) remain largely unknown. The goal of this study was to explore the role and potential mechanisms by which PMT contributes to subretinal fibrosis. Choroidal neovascularization (CNV) was induced by laser photocoagulation in transgenic mice with the collagen1α1-green fluorescent protein (Col1α1-GFP) reporter, and recombinant adeno-associated virus 2 (rAAV2)-mediated TGF-ß2 (rAAV2-TGF-ß2) was administered intravitreally to further induce PMT. Primary mouse choroidal GFP-positive pericytes were treated with TGF-ß2 in combination with siRNAs targeting Smad2/3, the Akt inhibitor MK2206 or the mTOR inhibitor rapamycin to examine cell proliferation, migration, and differentiation into myofibroblasts. The involvement of the Akt/mTOR pathway in PMT in subretinal fibrosis was further investigated in vivo. Intraocular TGF-ß2 overexpression induced GFP-positive pericyte infiltration and PMT in subretinal fibrosis, which was mimicked in vitro. Knockdown of Smad2/3 or inhibition of Akt/mTOR decreased cell proliferation, PMT and migration in primary mouse pericytes. Combined inhibition of Smad2/3 and mTOR showed synergistic effects on attenuating α-smooth muscle actin (α-SMA) expression and cell proliferation. In mice with laser-induced CNV, the administration of the Akt/mTOR inhibitors suppressed pericyte proliferation and alleviated the severity of subretinal fibrosis. Our results showed that PMT plays a pivotal role in subretinal fibrosis, which was induced by TGF-ß2 through the Smad2/3 and Akt/mTOR pathways. Thus, inhibiting PMT may be a novel strategy for the treatment of subretinal fibrosis.


Myofibroblasts , Pericytes , Animals , Fibrosis , Mice , Myofibroblasts/metabolism , Pericytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta2/metabolism
8.
Dis Model Mech ; 15(3)2022 03 01.
Article En | MEDLINE | ID: mdl-35230417

Bietti crystalline dystrophy (BCD) is an autosomal recessive inherited retinal disease, resulting in blindness in most patients. The etiology and development mechanism of it remain unclear. Given the defects in previous mouse models of BCD, we generated a new Cyp4v3-/- mouse model, using CRISPR/Cas9 technology, for investigating the pathogenesis of BCD. We estimated the ocular phenotypes by fundus imaging, optical coherence tomography (OCT) and full-field scotopic electroretinography, and investigated the histological features by Hematoxylin and Eosin staining, Oil Red O staining and immunofluorescence. This model effectively exhibited age-related progression that mimicked the human ocular phenotypes. Moreover, gas chromatography-mass spectrometry and RNA-seq analysis indicated that the defect of Cyp4v3 led to the abnormal lipid metabolism, inflammation activation and oxidative stress of retina. Notably, inflammation activation and oxidative stress could also promote the progression of BCD in light-induced retinal degeneration. In conclusion, our data provided evidence that we established a novel and more effective Cyp4v3 knockout preclinical mouse model for BCD, which served as a useful tool for evaluating the effect of drugs and gene therapy in vivo.


Corneal Dystrophies, Hereditary , Retinal Degeneration , Animals , Corneal Dystrophies, Hereditary/genetics , Disease Models, Animal , Humans , Mice , Mutation , Retinal Diseases , Tomography, Optical Coherence
9.
Int J Ophthalmol ; 14(9): 1334-1344, 2021.
Article En | MEDLINE | ID: mdl-34540608

AIM: To illustrate the underlying mechanism how prominin-1 (also known as Prom1) mutation contribute to progressive photoreceptor degeneration. METHODS: A CRISPR-mediated Prom1 knockout (Prom1-KO) mice model in the C57BL/6 was generated and the photoreceptor degeneration phenotypes by means of structural and functional tests were demonstrated. Immunohistochemistry and immunoblot analysis were performed to reveal the localization and quantity of related outer segment (OS) proteins. RESULTS: The Prom1-KO mice developed the photoreceptor degeneration phenotype including the decreased outer nuclear layer (ONL) thickness and compromised electroretinogram amplitude. Immunohistochemistry analysis revealed impaired trafficking of photoreceptor OS proteins. Immunoblot data demonstrated decreased photoreceptor OS proteins. CONCLUSION: Prom1 deprivation causes progressive photoreceptor degeneration. Prom1 is essential for maintaining normal trafficking and normal quantity of photoreceptor OS proteins. The new light is shed on the pathogenic mechanism underlying photoreceptor degeneration caused by Prom1 mutation.

10.
FASEB J ; 35(9): e21859, 2021 09.
Article En | MEDLINE | ID: mdl-34418172

Cyclic nucleotide-gated (CNG) channels are important mediators in the transduction pathways of rod and cone photoreceptors. Native CNG channels are heterotetramers composed of homologous A and B subunits. Biallelic mutations in CNGA1 or CNGB1 genes result in autosomal recessive retinitis pigmentosa (RP). To investigate the pathogenic mechanism of CNG channel-associated retinal degeneration, we developed a mouse model of CNGA1 knock-out using CRISPR/Cas9 technology. We observed progressive retinal thinning and a concomitant functional deficit in vivo as typical phenotypes for RP. Immunofluorescence and TUNEL staining showed progressive degeneration in rods and cones. Moreover, microglial activation and oxidative stress damage occurred in parallel. RNA-sequencing analysis of the retinae suggested down-regulated synaptic transmission and phototransduction as early as 9 days postnatal, possibly inducing later photoreceptor degeneration. In addition, the down-regulated PI3K-AKT-mTOR pathway indicated upregulation of autophagic process, and chaperone-mediated autophagy was further shown to coincide with the time course of photoreceptor death. Taken together, our studies add to a growing body of research exploring the mechanisms of photoreceptor death during RP progression and provide a novel CNGA1 knockout mouse model for potential development of therapies.


Cyclic Nucleotide-Gated Cation Channels/deficiency , Cyclic Nucleotide-Gated Cation Channels/genetics , Gene Deletion , Retinal Degeneration/genetics , Animals , Caspases , Chaperone-Mediated Autophagy , Female , Light , Mice , Mice, Knockout , Microglia , Neuroglia , Oxidative Stress/radiation effects , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , RNA-Seq , Retinal Degeneration/pathology , Retinal Pigment Epithelium , Transcriptome
11.
Front Neurosci ; 15: 610788, 2021.
Article En | MEDLINE | ID: mdl-34121982

Glaucoma, a neurodegenerative disease that leads to irreversible vision loss, is characterized by progressive loss of retinal ganglion cells (RGCs) and optic axons. To date, elevated intraocular pressure (IOP) has been recognized as the main phenotypic factor associated with glaucoma. However, some patients with normal IOP also have glaucomatous visual impairment and RGC loss. Unfortunately, the underlying mechanisms behind such cases remain unclear. Recent studies have suggested that retinal glia play significant roles in the initiation and progression of glaucoma. Multiple types of glial cells are activated in glaucoma. Microglia, for example, act as critical mediators that orchestrate the progression of neuroinflammation through pro-inflammatory cytokines. In contrast, macroglia (astrocytes and Müller cells) participate in retinal inflammatory responses as modulators and contribute to neuroprotection through the secretion of neurotrophic factors. Notably, research results have indicated that intricate interactions between microglia and macroglia might provide potential therapeutic targets for the prevention and treatment of glaucoma. In this review, we examine the specific roles of microglia and macroglia in open-angle glaucoma, including glaucoma in animal models, and analyze the interaction between these two cell types. In addition, we discuss potential treatment options based on the relationship between glial cells and neurons.

12.
Transl Vis Sci Technol ; 10(7): 25, 2021 06 01.
Article En | MEDLINE | ID: mdl-34144598

Purpose: This study aims to evaluate the pathogenesis of cone/cone-rod dystrophy (CoD/CoRD) caused by a cone-rod homeobox (CRX) mutation, which was identified in a Chinese family, through adeno-associated virus (AAV)-mediated overexpression of mutant CRX protein in the mouse retina. Methods: Comprehensive ophthalmologic examinations were performed for the pedigree members of a Chinese family with CoD/CoRD. Whole exome sequencing and Sanger sequencing were performed to determine the genetic cause of the disease. Furthermore, AAV vectors were used to construct AAV-CRX-mut-HA, which was transfected into mouse photoreceptor cells to clarify the pathogenesis of the mutant CRX. Results: Fundus photography and optical coherence tomography images displayed features that were consistent with CoD/CoRD, including macular atrophy and photoreceptor layer thinning. Electroretinogram analysis indicated an obvious decrease in photopic responses or both scotopic and photopic responses in affected individuals. A frameshift variant c.611delC (p.S204fs) in CRX was cosegregated with the disease in this family. AAV-CRX-mut-HA that subretinally injected into the C57BL/6 mice generally transfected the outer nuclear layer, leading to the loss of cone and rod photoreceptor cells, abnormal expression of CRX target genes, and a decrease in electroretinogram responses. Conclusions: AAV-mediated overexpression of CRX[S204fs] in the mouse retina led to a CoRD-like phenotype and showed the possible pathogenesis of the antimorphic CRX mutation. Translational Relevance: This study provides a modeling method to evaluate the pathogenesis of CoD/CoRD and other inherited retinal dystrophies caused by distinct gain-of-function mutations.


Cone-Rod Dystrophies , Animals , Dependovirus/genetics , Homeodomain Proteins , Mice , Mice, Inbred C57BL , Mutant Proteins , Retina , Trans-Activators
13.
J Craniomaxillofac Surg ; 49(6): 480-487, 2021 Jun.
Article En | MEDLINE | ID: mdl-33750636

The aim of this study was to classify the clinical feasibility and outcomes of open reduction treatment of old condylar head fractures (CHFs). This was a retrospective case series study of patients with old CHFs that were treated with open reduction and internal fixation, with anatomic reduction and sutured fixation of the articular disc. Preoperative and postoperative examinations were recorded and analyzed, including temporomandibular joint (TMJ) symptoms, occlusion, maximum interincisal opening (MIO), and mandibular deviation. Computed tomography (CT) was used to assess condylar morphology and position. Eleven patients with old CHFs were included (nine unilateral and two bilateral). The mean period from condylar fracture to operation was 8.9 months (ranging from 6 to 14 months). The mean follow-up period after surgery was 16.1 months (ranging from 12 to 22 months). At the end of follow-up period, no malocclusion was found, and the MIO had expanded considerably to 37.4 ± 3.8 mm. Postoperative CT showed that all fragments were properly reduced and the condyles were in the normal position. All patients showed apparently improved TMJ function, occlusion, and facial appearance. Our results showed that open reduction treatment could be an effective method for the treatment of old CHFs.


Mandibular Fractures , Adult , Follow-Up Studies , Fracture Fixation, Internal , Humans , Mandibular Condyle/diagnostic imaging , Mandibular Condyle/surgery , Mandibular Fractures/diagnostic imaging , Mandibular Fractures/surgery , Open Fracture Reduction , Retrospective Studies , Treatment Outcome
14.
Front Cell Dev Biol ; 9: 639851, 2021.
Article En | MEDLINE | ID: mdl-33681224

In age-related macular degeneration (AMD), one of the principal sources of vascular endothelial growth factor (VEGF) is retinal pigment epithelium (RPE) cells under hypoxia or oxidative stress. Solute carrier family 7 member 11 (SLC7A11), a key component of cystine/glutamate transporter, regulates the level of cellular lipid peroxidation, and restrains ferroptosis. In our study, we assessed the role of SLC7A11 in laser-induced choroidal neovascularization (CNV) and explored the underlying mechanism. We established a mouse model of CNV to detect the expression level of SLC7A11 and VEGF during disease progression. We found the expression of the SLC7A11 protein in RPE cells peaked at 3 days after laser treatment, which was correlated with the expression of VEGF. Intraperitoneal injection of SLC7A11 inhibitor expanded the area of CNV. We examined functional proteins related to oxidative stress and Fe2+ and found laser-induced ferroptosis accompanied by increased Fe2+ content and GPX4 expression in the RPE-choroidal complex after laser treatment. We verified the expression of SLC7A11 in the ARPE19 cell line and the effects of its inhibitors on cell viability and lipid peroxidation in vitro. Application of SLC7A11 inhibitor and SLC7A11 knockdown increased the level of lipid peroxidation and reduced the cell viability of ARPE19 which can be rescued by ferroptosis inhibitors ferrostatin-1 (Fer-1) and liproxstatin-1 (Lip-1). Conversely, SLC7A11 overexpression induced resistance to erastin or RSL3-induced ferroptosis. Moreover, we tested the possible regulatory transcription factor NF-E2-related factor 2 (NRF2) of SLC7A11 by Western blot. Knock-down of NRF2 decreased the expression of SLC7A11. Our study suggests that SLC7A11 plays a key role in the laser-induced CNV model by protecting RPE cells from ferroptosis. SLC7A11 provides a new therapeutic target for neovascular AMD patients.

15.
Biomed Res Int ; 2020: 1454396, 2020.
Article En | MEDLINE | ID: mdl-33299856

BACKGROUND: Combined aerobic and resistance training has been demonstrated to benefit glycemic control and reverse nonalcoholic fatty liver disease in childhood obesity. General control nonderepressible 2 (GCN2) deficiency has been reported to attenuate hepatic steatosis and insulin resistance. However, whether GCN2 impacts the positive effects of combined aerobic and resistance exercise remains unknown. OBJECTIVES: To investigate whether combined aerobic and resistance exercise improves hepatic steatosis and glucose intolerance and the role GCN2 plays in mediating the metabolic regulation of exercise. METHODS: Wild-type (WT) and GCN2 knockout (GCN2KO) mice were fed a high-fat diet (HFD) for 25 weeks. The WT and GCN2KO mice performed exercise (treadmill running + ladder climbing) during the last eight weeks. Their body and liver weights, their triglyceride content, and their levels of aspartate transaminase (AST), alanine transaminase (ALT), and blood glucose were measured, and the expressions of proteins involved in the GCN2/eIF2α/ATF4 pathway and the glucolipid metabolism-related proteins (e.g., p-AMPK, SIRT1, PPARα, PGC-1α, GLUT4, and p-GSK-3ß) were determined. RESULTS: The body weight of WT and GCN2KO mice continued to increase until the end of the experiment. The liver weights, hepatic triglyceride content, and AST and ALT levels of the exercised mice were significantly reduced compared to those of the sedentary mice. Exercise improved blood glucose levels and glucose clearance ability in the WT mice, but the glucose intolerance of GCN2KO mice was not improved. Exercise increased PGC-1α, GLUT4, and p-GSK-3ß expressions in the WT rather than the GCN2KO mice. Interestingly however, exercise-trained GCN2KO mice were better protected against hepatic steatosis with downregulated expressions of p-eIF2α and ATF4, upregulated expressions of p-AMPK and SIRT1, and the presence of PPARα in the liver, compared to the exercised WT mice. CONCLUSION: Combined aerobic and resistance exercise had positive effects on hepatic steatosis and the control of glucose intolerance. GCN2 was found to be necessary for exercise-induced improved glucose intolerance. However, the better efficacy in improving hepatic steatosis by exercise in the GCN2-deficient mice enhanced liver lipid metabolism, at least partially, via the AMPK/SIRT1/PPARα pathway.


Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Physical Conditioning, Animal , Protein Serine-Threonine Kinases/deficiency , Activating Transcription Factor 4/metabolism , Adenylate Kinase/metabolism , Animals , Diet, High-Fat , Eukaryotic Initiation Factor-2/metabolism , Glycolipids/metabolism , Lipid Metabolism , Liver/metabolism , Liver/pathology , Liver/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Organ Size , PPAR alpha/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Sirtuin 1/metabolism , Triglycerides/metabolism
16.
Free Radic Biol Med ; 158: 32-43, 2020 10.
Article En | MEDLINE | ID: mdl-32679366

BACKGROUNDS: Photoreceptor degeneration underlies various retinal disorders that lead to vision impairment. Currently, no effective medication is available to rescue photoreceptors under disease conditions. Elucidation of the molecular pathways involved in photoreceptor degeneration is a prerequisite for the rational design of therapeutic interventions. Photoreceptors are among the most energy-demanding tissues that require highly active oxidative phosphorylation. Therefore, disruption of metabolic support to photoreceptors results in a redox imbalance and subsequent cell death. We hypothesize that the redox regulatory pathway could be a potential therapeutic target to rescue photoreceptors under disease conditions. METHODS: Experimental retinal detachment was induced in mice. A murine photoreceptor-derived 661w cell line treated with H2O2 was employed as an in vitro model to study the cellular response to oxidative stress. The expression and functional role of xCT, an upstream regulator of redox homeostasis, was assessed in vivo and in vitro. An xCT expression vector was constructed for an in vivo study to evaluate the therapeutic potential of this molecule. RESULTS: xCT expression was upregulated in detached retina and H2O2-stimulated 661w cells compared to the control cells. Pharmacological inhibition of xCT by sulfasalazine (SAS) promoted photoreceptor degeneration after retinal detachment and 661w cell death upon H2O2 treatment. Additionally, SAS treatment induced reactive oxidative species (ROS) accumulation, glutathione (GSH) depletion, and glutamate release in 661w cells. In contrast, xCT overexpression via viral infection protected photoreceptors from degeneration after retinal detachment. CONCLUSION: We conclude that xCT expression is upregulated in photoreceptors after retinal detachment and plays a neuroprotective role in preserving photoreceptors. Mechanistically, xCT promotes cellular homeostasis by regulating intracellular ROS and GSH levels, which are critical to photoreceptor survival after retinal detachment. Collectively, our findings identify xCT as a potential therapeutic target for protection of photoreceptors under disease conditions.


Retinal Degeneration , Retinal Detachment , Animals , Homeostasis , Hydrogen Peroxide , Mice , Oxidation-Reduction , Photoreceptor Cells, Vertebrate , Retinal Degeneration/drug therapy , Retinal Degeneration/genetics
17.
Invest Ophthalmol Vis Sci ; 61(8): 31, 2020 07 01.
Article En | MEDLINE | ID: mdl-32697303

Purpose: Gasdermin D (GSDMD) is crucial in neuronal pyroptosis. GSDMD-N and GSDMD-C are two subdomains of the protein GSDMD. GSDMD-N is an executor of pyroptosis, and GSDMD-C has an inhibitory effect on pyroptotic cell death. This study evaluated the role of GSDMD in photoreceptor cell pyroptosis caused by retinal detachment (RD). Methods: RD models were established in rats, and GSDMD cleavage was detected by western blotting. The morphology of photoreceptors was assessed by transmission electron microscopy. Some rats were given subretinal injections of recombinant adeno-associated virus 2/8 (rAAV2/8)-GSDMD-C before RD surgery. We documented the expression of caspase-1 and GSDMD-N in retinas by western blot. Levels of IL-1ß, TNF-α, and monocyte chemoattractant protein-1 (MCP-1) were detected by quantitative RT-PCR. The membrane integrity of photoreceptors was evaluated by TOTO-3 iodide staining. Retinal function was measured by electroretinography, and the thickness of the outer nuclear layer was also recorded. We measured the activation of glial fibrillary acidic protein (GFAP), F4/80, and ionized calcium binding adaptor molecule 1 (Iba-1) by immunofluorescence. Results: The cleavage of GSDMD peaked at 1 day after RD. The administration of rAAV2/8-GSDMD-C reduced the pyroptosis and subsequent apoptosis of photoreceptors and preserved the retinal function after RD. Expression of IL-1, TNF-α, and MCP-1 was decreased in the rAAV2/8-GSDMD-C group. In addition, the activation of GFAP, Iba-1, and F4/80 in retinas was alleviated by administering rAAV2/8-GSDMD-C after RD. Conclusions: GSDMD participates in the pyroptosis of photoreceptor after RD. Overexpression of GSDMD-C may block GSDMD cleavage and attenuate photoreceptor degeneration.


Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Pyroptosis , Retina , Retinal Detachment/metabolism , Animals , Apoptosis , Chemokine CCL2/metabolism , Disease Models, Animal , Electroretinography/methods , Interleukin-1beta/metabolism , Rats , Retina/diagnostic imaging , Retina/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
18.
J Ocul Pharmacol Ther ; 36(8): 618-628, 2020 10.
Article En | MEDLINE | ID: mdl-32552228

Purpose: To explore the effects of 3-methyladenine (3-MA), a selective inhibitor of phosphatidylinositol-3-kinase (PI3K), on experimental subretinal fibrosis (SRF) in mice. Methods: The SRF mouse model was established by 532 nm laser photocoagulation at each fundus of mice on day 0. 3-MA was administered every 2 days from day 0 to 35. Immunofluorescence of choroidal flat mounts was performed to evaluate the size of SRF area, local macrophages, and polarization, respectively. Besides, Western blot analysis was carried out to assess the expression levels of macrophage polarization-related genes, Arg-1, Ym-1, and transforming growth factor-ß2 (TGF-ß2). Co-culture and migration experiments were used to demonstrate the inhibitory effect of 3-MA on fibroblasts. The gene knockout and Western blot analysis were used to explore the signal pathways related to macrophage polarization. Results: Compared with the control group, the 3-MA-treated group showed significantly less size of SRF area. 3-MA treatment reduced both circulating and local macrophages, and counteracted M2 polarization. Moreover, 3-MA inhibited fibroblast recruitment. Mechanistically, we proved that 3-MA inhibits macrophage M2 polarization by suppressing PI3K/Akt signal pathway rather than the PI3K-autophagy-related signal pathway. Conclusions: 3-MA exerts antifibrotic effects on experimental SRF by targeting circulating and local macrophages and M2 polarization, through PI3K/Akt signal pathway. These results support the potential use of 3-MA as a new therapeutic modality for SRF associated with neovascular age-related macular degeneration.


Adenine/analogs & derivatives , Fibrosis/drug therapy , Macrophages/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Adenine/administration & dosage , Adenine/pharmacology , Animals , Choroid Diseases/metabolism , Choroid Diseases/pathology , Fibroblasts/drug effects , Fluorescent Antibody Technique/methods , Macrophages/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/pathology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Signal Transduction/drug effects
19.
Biosci Trends ; 14(2): 115-122, 2020 May 21.
Article En | MEDLINE | ID: mdl-32238672

Dimethylarginine dimethylaminohydrolase-1 (DDAH1) maintains nitric oxide (NO) bioavailability by degrading asymmetric dimethylarginine (ADMA), which is an endogenous inhibitor of nitric oxide synthase (NOS). It has been well established that DDAH1 and exercise play crucial roles in promoting cardiac angiogenesis under pathological conditions. However, the role of DDAH1 in exercise-induced cardiac angiogenesis remains unclear. In this study, we focused on the change in DDAH1 in response to moderate exercise and the underlying mechanism of exercise-induced cardiac angiogenesis. Eight-week-old male DDAH1 global knockout (KO) mice and DDAH1flox/flox mice (wild-type) were randomly divided into sedentary groups (control) and swimming groups (exercise). After eight weeks of swimming at five days per week, all the mice were anesthetized and sacrificed. Histological examination and Western blot analysis were performed. There were low levels of myocardial capillaries in DDAH1 KO mice under control and exercise conditions. Notably, exercise elevated DDAH1 protein expression, as observed by Western blot analysis. The common cardiac angiogenesis biomarkers vascular endothelial growth factor (VEGF) and Caveolin-1 were increased during exercise. A significant difference in VEGF was observed between the DDAH1 KO and wild-type groups. Similarly, increased Caveolin-1 expression was abrogated in DDAH1 KO mice. Furthermore, we tested the R-Ras/AKT/GSK3ß signaling pathway to study the underlying molecular mechanism. DDAH1 may regulate the R-Ras/AKT/GSK3ß pathway due to distinct protein changes in this pathway in the DDAH1 KO and wild-type groups. Our findings suggest that DDAH1 plays an important role in exercise-induced cardiac angiogenesis by regulating the R-Ras/AKT/GSK3ßsignaling pathway.


Amidohydrolases/metabolism , Coronary Vessels/growth & development , Heart/physiology , Neovascularization, Physiologic , Physical Conditioning, Animal/physiology , Amidohydrolases/genetics , Animals , Arginine/analogs & derivatives , Arginine/metabolism , Behavior, Animal , Glycogen Synthase Kinase 3 beta/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Nitric Oxide/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sedentary Behavior , Signal Transduction/physiology , ras Proteins/metabolism
20.
Exp Eye Res ; 193: 107949, 2020 04.
Article En | MEDLINE | ID: mdl-32006561

Retinal detachment (RD) results in disruption of retinal physiology and visual function. Although surgical intervention has been well-developed to restore the retinal anatomic structure, post-op progression of visual function decline is prominent in a large proportion of patients. Therefore, the establishment of a disease model that accurately mimics RD pathogenesis is crucial to mechanistic study and drug screening. General protocols to induce RD in mice are frequently associated with complications leading to model instability and reduced reproducibility. In this study, we established a stable and reproducible mice RD model with a detached area of over 90% and rare complications. Briefly, the modified method was realized by vitreous humor extraction to reduce intraocular pressure, followed by directly-visible hyaluronic acid injection into subretinal space. The detachment of retina was confirmed by fundus photography, and progressive thinning of the outer nuclear layer (ONL) was determined by HE staining. Apoptotic signals were prominent in the ONL. Consistently, visual function was significantly compromised as determined by ERG. Moreover, retinal vasculature appeared to remodel and acquired winding, twisted and dilated structures illustrated by 3D reconstruction. In addition, activation of Müller cells and microglia, and infiltration of blood-derived macrophages were detected locally. Collectively, we have established a modified protocol to model RD with increased stability, reproducibility and fewer complications, and 3D high-resolution imaging and reconstruction of vasculature could provide new tools to evaluate this model.


Electroretinography/methods , Imaging, Three-Dimensional/methods , Retina/diagnostic imaging , Retinal Detachment/diagnosis , Retinal Vessels/diagnostic imaging , Tomography, Optical Coherence/methods , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL
...