Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1870(1): 159566, 2024 Sep 29.
Article in English | MEDLINE | ID: mdl-39349136

ABSTRACT

The translocator protein, TSPO, has been implicated in a wide range of cellular processes exerted from its position in the outer mitochondrial membrane from where it influences lipid metabolism and mitochondrial oxidative activity. Understanding how this protein regulates a profusion of processes requires further elucidation and to that end we have examined lipid metabolism and used an RNAseq strategy to compare transcript abundance in wildtype and Tspo knockout (KO) mouse liver. The levels of cholesterol, triglyceride and phospholipid were significantly elevated in the KO mouse liver. The expression of cholesterol homeostasis genes was markedly downregulated. Determination of the differential expression revealed that many genes were either up- or downregulated in the KO animals. However, a striking observation within the results was a decrease of transcripts for protein degradation proteins in KO animals while protease inhibitors were enriched. When the entire abundance data-set was analysed with CEMiTool, and revealed a module of proteins that were under-represented in the KO animals. These could subsequently be formed into a network comprising three interlinked clusters at the centre of which were proteins of cytoplasmic ribosomes with gene ontology terms suggesting impairment to translation. The largest cluster was dominated by proteins of lipid metabolism but also contained disparate systems of iron metabolism and behaviour. The third cluster was dominated by proteins of the electron transport chain and oxidative phosphorylation. These findings suggest that TSPO contributes to lipid metabolism, detoxification of active oxygen species and oxidative phosphorylation, and regulates mitochondrial retrograde signalling.

2.
Cell ; 186(2): 382-397.e24, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36669473

ABSTRACT

Blood and lymphatic vessels form a versatile transport network and provide inductive signals to regulate tissue-specific functions. Blood vessels in bone regulate osteogenesis and hematopoiesis, but current dogma suggests that bone lacks lymphatic vessels. Here, by combining high-resolution light-sheet imaging and cell-specific mouse genetics, we demonstrate presence of lymphatic vessels in mouse and human bones. We find that lymphatic vessels in bone expand during genotoxic stress. VEGF-C/VEGFR-3 signaling and genotoxic stress-induced IL6 drive lymphangiogenesis in bones. During lymphangiogenesis, secretion of CXCL12 from proliferating lymphatic endothelial cells is critical for hematopoietic and bone regeneration. Moreover, lymphangiocrine CXCL12 triggers expansion of mature Myh11+ CXCR4+ pericytes, which differentiate into bone cells and contribute to bone and hematopoietic regeneration. In aged animals, such expansion of lymphatic vessels and Myh11-positive cells in response to genotoxic stress is impaired. These data suggest lymphangiogenesis as a therapeutic avenue to stimulate hematopoietic and bone regeneration.


Subject(s)
Bone Regeneration , Lymphatic Vessels , Aged , Animals , Humans , Mice , Endothelial Cells , Lymphangiogenesis
3.
Exp Eye Res ; 208: 108625, 2021 07.
Article in English | MEDLINE | ID: mdl-34022174

ABSTRACT

Age-related Macular Degeneration (AMD) is a major cause of sight impairment in the elderly with complex aetiology involving genetics and environment and with limited therapeutic options which have limited efficacy. We have previously shown in a mouse-model of the condition, induced by feeding a high fat diet, that adverse effects of the diet can be reversed by co-administration of the TSPO activator, etifoxine. We extend those observations showing improvements in retinal pigment epithelial (RPE) cells with decreased lipids and enhanced expression of cholesterol metabolism and transport enzymes. Further, etifoxine decreased levels of reactive oxygen species (ROS) in RPE and inflammatory cytokines in RPE and serum. With respect to gut microbiome, we found that organisms abundant in the high fat condition (e.g. in the genus Anaerotruncus and Oscillospira) and implicated in AMD, were much less abundant after etifoxine treatment. The changes in gut flora were associated with the predicted production of metabolites of benefit to the retina including tryptophan and other amino acids and taurine, an essential component of the retina necessary to counteract ROS. These novel observations strengthen earlier conclusions that the mechanisms behind improvements in etifoxine-induced retinal physiology involve an interaction between effects on the host and the gut microbiome.


Subject(s)
Cholesterol/metabolism , Lipid Metabolism , Macular Degeneration/metabolism , Oxidative Stress/physiology , Receptors, GABA/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Homeostasis , Ligands , Macular Degeneration/pathology , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/pathology
4.
Article in English | MEDLINE | ID: mdl-33771709

ABSTRACT

Age-related macular degeneration (AMD) is the most common cause of visual disorder in aged people and may lead to complete blindness with ageing. The major clinical feature of AMD is the presence of cholesterol enriched deposits underneath the retinal pigment epithelium (RPE) cells. The deposits can induce oxidative stress and inflammation. It has been suggested that abnormal cholesterol homeostasis contributes to the pathogenesis of AMD. However, the functional role of defective cholesterol homeostasis in AMD remains elusive. STARD proteins are a family of proteins that contain a steroidogenic acute regulatory protein-related lipid transfer domain. There are fifteen STARD proteins in mammals and some, such as STARD3, are responsible for cholesterol trafficking. Previously there was no study of STARD proteins in retinal cholesterol metabolism and trafficking. Here we examined expression of the Stard3 gene in mouse retinal and RPE cells at ages of 2 and 20 months. We found that expression of Stard 3 gene transcripts in both mouse RPE and retina was significantly decreased at age of 20 months when compared to that of age 2 months old. We created a stable ARPE-19 cell line overexpressing STARD3 and found this resulted in increased cholesterol efflux, reduced accumulation of intracellular oxidized LDL, increased antioxidant capacity and lower levels of inflammatory cytokines. The data suggested that STARD3 is a potential target for AMD through promoting the removal of intracellular cholesterol and slowing the disease progression.


Subject(s)
Lipoproteins, LDL/pharmacology , Membrane Proteins/genetics , Oxidative Stress/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Animals , Cell Line , Gene Expression , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice
5.
Exp Eye Res ; 201: 108291, 2020 12.
Article in English | MEDLINE | ID: mdl-33049273

ABSTRACT

Retinitis pigmentosa (RP) is a collection of heterogenous genetic retinal disorders resulting in cumulative retinal deterioration involving progressive loss of photoreceptors and eventually in total blindness. Oxidative stress plays a central role in this photoreceptor loss. Gypenosides (Gyp) are the main functional component isolated from the climbing vine Gynostemma pentaphyllum and have been shown to defend cells against the effects of oxidative stress and inflammation, providing protection in experimentally-induced optic neuritis. The zebrafish model has been used to investigate a range of human diseases. Previously we reported early retinal degeneration in a mutant zebrafish line carrying a point-nonsense mutation in the retinitis pigmentosa GTPase regulator interacting protein 1 (rpgrip1) gene that is mutated in RP patients. The current study investigated the potential protective effects of Gyp against photoreceptor degeneration in the Rpgrip1 deleted zebrafish. Rpgrip1 mutant zebrafish were treated with 5 µg/ml of Gyp in E3 medium from 6 h post fertilization (hpf) till 1 month post fertilization (mpf). Rpgrip1 mutant zebrafish treated with 5 µg/ml of Gyp showed a significant decrease by 68.41% (p = 0.0002) in photoreceptor cell death compared to that of untreated mutant zebrafish. Expression of antioxidant genes catalase, sod1, sod2, gpx1, gclm, nqo-1 and nrf-2 was significantly decreased in rpgrip1 mutant zebrafish eyes by 61.51%, 77.40%, 60.11%, 81.17%, 72.07%, 78.95% and 85.42% (all p < 0.0001), respectively, when compared to that of wildtype zebrafish; superoxide dismutase and catalase activities, and glutathione levels in rpgrip1 mutant zebrafish eyes were significantly decreased by 87.21%, 21.55% and 96.51% (all p < 0.0001), respectively. There were marked increases in the production of reactive oxygen species (ROS) and malondialdehyde (MDA) by 2738.73% and 510.69% (all p < 0.0001), respectively, in rpgrip1 mutant zebrafish eyes; expression of pro-inflammatory cytokines IL-1ß, IL-6 and TNF-α was also significantly increased by 150.11%, 267.79% and 190.72% (all p < 0.0001), respectively, in rpgrip1 mutant zebrafish eyes, compared to that of wildtype zebrafish. Treatment with Gyp significantly counteracted these effects. This study indicates that Gyp has a potential role in the treatment of RP.


Subject(s)
Oxidative Stress , Photoreceptor Cells, Invertebrate/drug effects , Retina/drug effects , Retinitis Pigmentosa/drug therapy , Animals , Gynostemma , Immunohistochemistry , Photoreceptor Cells, Invertebrate/metabolism , Photoreceptor Cells, Invertebrate/pathology , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Rhodopsin/metabolism , Zebrafish
6.
Biochem Pharmacol ; 180: 114151, 2020 10.
Article in English | MEDLINE | ID: mdl-32679124

ABSTRACT

Obesity is intimately associated with diet and dysbiosis of gut microorganisms but anxiolytics, widely used in treatment of psychiatric conditions, frequently result in weight gain and associated metabolic disorders. We are interested in effects of the anxiolytic etifoxine, which has not been studied with respect to weight gain or effects on gut microorganisms. Here we induced obesity in mice by feeding a high-fat diet but found that intraperitoneal administration of etifoxine resulted in weight loss and decreased serum cholesterol and triglycerides. Obese mice had increased hepatic transcripts associated with lipid metabolism (cyp7a1, cyp27a1, abcg1 and LXRα) and inflammatory factors (TNFα and IL18) but these effects were reversed after etifoxine treatment other than cyp7a1. Taxonomic profiles of the organisms from the caecum were generated by 16S rRNA gene sequencing and Obese and etifoxine mice show differences by diversity metrics, Differential Abundance and functional metagenomics. Organisms in genus Oscillospira and genera from Lachnospiraceae family and Clostridiales order are higher in Control than Obese and at intermediate levels with etifoxine treatment. With respect to community metabolic potential, etifoxine mice have characteristics similar to Control and particularly with respect to metabolism of butanoate, sphingolipid, lipid biosynthesis and xenobiotic metabolism. We suggest mechanisms where-by etifoxine influences processes of host, such as on bile acid synthesis, and microbiota, such as signalling from production of butanoate and sphingosine, resulting in decreased cholesterol, lipids and inflammatory factors. We speculate that the indirect effect of etifoxine on microbial composition is mediated by microbial ß-glucuronidases that metabolise excreted etifoxine glucuronides.


Subject(s)
Colon/drug effects , Gastrointestinal Microbiome/drug effects , Obesity/drug therapy , Oxazines/pharmacology , Oxazines/therapeutic use , Weight Gain/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Colon/microbiology , Colon/physiology , Diet, High-Fat/adverse effects , Disease Models, Animal , Gastrointestinal Microbiome/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/physiopathology , Weight Gain/physiology , Weight Loss/drug effects , Weight Loss/physiology
7.
Exp Cell Res ; 392(1): 112023, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32325079

ABSTRACT

Diabetic retinopathy (DR) is a diabetes-associated complication characterized by irreversible deterioration of the microvessels within the retina, leading subsequently to severe retinal damage and vision loss. Vitamin D (VITD), a steroid hormone, plays multiple physiological functions in cellular homeostasis. Deficiency of VITD has been suggested to be associated with DR. To study the potential protective function of VITD in DR, high-glucose-treated ARPE-19 cells and STZ-induced diabetic mice were used as in vitro and in vivo models. The protective effects of VITD were assessed based on the changes of expression of antioxidant enzymes and cytokines in high-glucose-treated retinal pigment epithelial (RPE) cells and in the retina and RPE of diabetic and VITD-treated diabetic mice. The present study demonstrated that exposure to a high level of glucose caused upregulation of pro-inflammatory cytokines and a decrease in anti-oxidant enzyme expression in both in vitro and in vivo models. VITD treatment increased cell viability, reduced reactive oxygen species (ROS) production and caspase-3/7 activities in high-glucose-treated RPE cells. Our data suggest that VITD can protect the retina and RPE from high-glucose-induced oxidative damage and inflammation.


Subject(s)
Cytoprotection/drug effects , Epithelial Cells/drug effects , Glucose/adverse effects , Retinal Pigment Epithelium/drug effects , Vitamin D/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/chemically induced , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/pathology , Diabetic Retinopathy/pathology , Diabetic Retinopathy/prevention & control , Dose-Response Relationship, Drug , Epithelial Cells/physiology , Glucose/pharmacology , Humans , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Protective Agents/pharmacology , Protective Agents/therapeutic use , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/physiology , Streptozocin , Vitamin D/therapeutic use
8.
Exp Eye Res ; 191: 107931, 2020 02.
Article in English | MEDLINE | ID: mdl-31931003

ABSTRACT

Age-related macular degeneration (AMD) is a predominant cause of visual deficit in aged population. Abnormal accumulation of cholesterol, including oxidized low-density lipoprotein (oxLDL), underneath the retinal pigment epithelium (RPE) cells contributes to the development of AMD. Gypenosides (Gyp) are glycosides extracted from Gynostemma pentaphyllum and have demonstrated protective effects against inflammation and oxidative stress. To determine the therapeutic potential of Gyp for AMD, we investigated its effect on cholesterol trafficking and metabolism and assessed the protective function of Gyp against oxLDL-induced damage in RPE cells. Cholesterol efflux to high-density lipoprotein (HDL) and human serum was significantly increased in RPE cells treated with Gyp when compared to untreated control cells. Expression of cholesterol metabolism (CYP27A1, CYP46A1) and trafficking (TSPO, ABCA1 and ABCG1) genes was also markedly increased in Gyp-treated RPE cells. OxLDL-treated RPE cells had significantly increased cholesterol accumulation and lipid droplet formation. There were marked increases in reactive oxygen species (ROS) generation and proinflammatory cytokines via NF-κB activation in RPE cells treated with oxLDL, while incubation with Gyp rectified these changes. These findings provide pharmacological evidence that Gyp has the potential to treat patients with early onset AMD by promoting cellular cholesterol removal from RPE cells and inhibiting inflammation and oxidative stress.


Subject(s)
Cholesterol/metabolism , Inflammation/metabolism , Lipoproteins, LDL/metabolism , Retinal Pigment Epithelium/drug effects , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Blotting, Western , Cell Line , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol 24-Hydroxylase/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/physiology , Gynostemma/chemistry , Humans , NF-kappa B/metabolism , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Receptors, GABA/metabolism , Retinal Pigment Epithelium/metabolism
9.
Antioxidants (Basel) ; 8(9)2019 Aug 24.
Article in English | MEDLINE | ID: mdl-31450606

ABSTRACT

Age-related macular degeneration (AMD), the most common visual disorder in elderly people, is characterized by the formation of deposits beneath the retinal pigment epithelium (RPE) and by dysfunction of RPE and photoreceptor cells. The biologically active form of vitamin D, 1,25-(OH)2D3 (VITD), is categorized as a multifunctional steroid hormone that modulates many transcriptional processes of different genes and is involved in a broad range of cellular functions. Epidemiological and genetic association studies demonstrate that VITD may have a protective role in AMD, while single nucleotide polymorphisms in the vitamin D metabolism gene (CYP24A1) increase the risk of AMD. However, the functional mechanisms of VITD in AMD are not fully understood. In the current study, we investigated the impact of VITD on H2O2-induced oxidative stress and inflammation in human RPE cells. We demonstrate that exposure to H2O2 caused significantly reduced cell viability, increased production of reactive oxygen species (ROS), lowered expression of antioxidant enzymes and enhanced inflammation. VITD exposure notably counteracted the above H2O2-induced effects. Our data suggest that VITD protects the RPE from oxidative damage and elucidate molecular mechanisms of VITD deficiency in the development of AMD.

10.
Int J Mol Sci ; 20(6)2019 Mar 19.
Article in English | MEDLINE | ID: mdl-30893912

ABSTRACT

Age-related macular degeneration is the main cause of vision loss in the aged population worldwide. Drusen, extracellular lesions formed underneath the retinal pigment epithelial (RPE) cells, are a clinical feature of AMD and associated with AMD progression. RPE cells support photoreceptor function by providing nutrition, phagocytosing outer segments and removing metabolic waste. Dysfunction and death of RPE cells are early features of AMD. The translocator protein, TSPO, plays an important role in RPE cholesterol efflux and loss of TSPO results in increased intracellular lipid accumulation and reactive oxygen species (ROS) production. This study aimed to investigate the impact of TSPO knockout on RPE cellular metabolism by identifying the metabolic differences between wildtype and knockout RPE cells, with or without treatment with oxidized low density lipoprotein (oxLDL). Using liquid chromatography mass spectrometry (LC/MS), we differentiated several metabolic pathways among wildtype and knockout cells. Lipids amongst other intracellular metabolites were the most influenced by loss of TSPO and/or oxLDL treatment. Glucose, amino acid and nucleotide metabolism was also affected. TSPO deletion led to up-regulation of fatty acids and glycerophospholipids, which in turn possibly affected the cell membrane fluidity and stability. Higher levels of glutathione disulphide (GSSG) were found in TSPO knockout RPE cells, suggesting TSPO regulates mitochondrial-mediated oxidative stress. These data provide biochemical insights into TSPO-associated function in RPE cells and may shed light on disease mechanisms in AMD.


Subject(s)
Epithelial Cells/metabolism , Gene Deletion , Metabolomics , Receptors, GABA/genetics , Retinal Pigment Epithelium/cytology , Cell Line , Discriminant Analysis , Epithelial Cells/drug effects , Glucose/metabolism , Glutathione Disulfide/metabolism , Humans , Lipid Metabolism/drug effects , Lipoproteins, LDL/pharmacology , Metabolome/drug effects , Nucleotides/metabolism , Oxidative Stress/drug effects , Principal Component Analysis , Receptors, GABA/metabolism
11.
Int J Mol Sci ; 19(12)2018 Nov 24.
Article in English | MEDLINE | ID: mdl-30477223

ABSTRACT

Choroidal endothelial cells supply oxygen and nutrients to retinal pigment epithelial (RPE) cells and photoreceptors, recycle metabolites, and dispose of metabolic waste through the choroidal blood circulation. Death of the endothelial cells of the choroid may cause abnormal deposits including unesterified and esterified cholesterol beneath RPE cells and within Bruch's membrane that contribute to the progression of age-related macular degeneration (AMD), the most prevalent cause of blindness in older people. Translocator protein (TSPO) is a cholesterol-binding protein that is involved in mitochondrial cholesterol transport and other cellular functions. We have investigated the role of TSPO in choroidal endothelial cells. Immunocytochemistry showed that TSPO was localized to the mitochondria of choroidal endothelial cells. Choroidal endothelial cells exposed to TSPO ligands (Etifoxine or XBD-173) had significantly increased cholesterol efflux, higher expression of cholesterol homeostasis genes (LXRα, CYP27A1, CYP46A1, ABCA1 and ABCG1), and reduced biosynthesis of cholesterol and phospholipids from [14C]acetate, when compared to untreated controls. Treatment with TSPO ligands also resulted in reduced production of reactive oxygen species (ROS), increased antioxidant capacity, and reduced release of pro-inflammatory cytokines (IL-1ß, IL-6, TNF-α and VEGF) induced by oxidized LDL. These data suggest TSPO ligands may offer promise for the treatment of AMD.


Subject(s)
Cholesterol/metabolism , Choroid/drug effects , Lipoproteins, LDL/antagonists & inhibitors , Oxazines/pharmacology , Purines/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Animals , Biological Transport/drug effects , Cell Line , Choroid/cytology , Choroid/metabolism , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation , Humans , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Ligands , Lipoproteins, LDL/pharmacology , Liver X Receptors/genetics , Liver X Receptors/metabolism , Macaca mulatta , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Phospholipids/antagonists & inhibitors , Phospholipids/biosynthesis , Reactive Oxygen Species/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Exp Eye Res ; 175: 103-114, 2018 10.
Article in English | MEDLINE | ID: mdl-29928899

ABSTRACT

Acrylamide (ACR) is a water-soluble chemical used widely in industry, which can be formed in tobacco smoke and in starchy foods cooked at high temperatures. ACR is considered to be a neurotoxin, genotoxin and carcinotoxin. Previous studies reported that ACR-exposed workers and experimental animals exhibited visual function defects, although the underlying mechanisms have not been elucidated. In this study, we found that zebrafish embryos exposed to 1 mM and 2 mM ACR showed significantly increased reactive oxygen species (ROS), decreased expression of the antioxidant genes Sod1, Sod2, Catalase, Gpx1 and Nrf2, reduced activity of superoxide dismutase (SOD) and catalase, and elevated malondialdehyde (MDA), compared with control embryos. ACR exposure caused loss of both rod and cone photoreceptor cells through Caspase-3-dependent apoptotis. When embryos were simultaneously exposed to ACR and the natural antioxidative substance carnosic acid (CA), the presence of the latter (10 µM) markedly counteracted the above ACR-induced toxic effects. Our data suggest that CA can protect photoreceptor cells against ACR-induced oxidative damage and has a potential for neuroprotection of visual function in humans exposed to ACR.


Subject(s)
Abietanes/pharmacology , Acrylamide/toxicity , Antioxidants/pharmacology , Embryo, Nonmammalian/drug effects , Oxidative Stress/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Zebrafish/embryology , Animals , Catalase/metabolism , Cell Survival/physiology , Embryo, Nonmammalian/metabolism , Glutathione Peroxidase/metabolism , NF-E2-Related Factor 2/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism , Zebrafish Proteins/metabolism , Glutathione Peroxidase GPX1
13.
Oncotarget ; 9(33): 23183-23197, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29796181

ABSTRACT

Ciliopathies are a group of genetically heterogeneous disorders, characterized by defects in cilia genesis or maintenance. Mutations in the RPGR gene and its interacting partners, RPGRIP1 and RPGRIP1L, cause ciliopathies, but the function of their proteins remains unclear. Here we show that knockdown (KD) of RPGR, RPGRIP1 or RPGRIP1L in hTERT-RPE1 cells results in abnormal actin cytoskeleton organization. The actin cytoskeleton rearrangement is regulated by the small GTPase RhoA via the planar cell polarity (PCP) pathway. RhoA activity was upregulated in the absence of RPGR, RPGRIP1 or RPGRIP1L proteins. In RPGR, RPGRIP1 or RPGRIP1L KD cells, we observed increased levels of DVl2 and DVl3 proteins, the core components of the PCP pathway, due to impaired proteasomal activity. RPGR, RPGRIP1 or RPGRIP1L KD cells treated with thapsigargin (TG), an inhibitor of sarcoendoplasmic reticulum Ca2+- ATPases, showed impaired store-operated Ca2+ entry (SOCE), which is mediated by STIM1 and Orai1 proteins. STIM1 was not localized to the ER-PM junction upon ER store depletion in RPGR, RPGRIP1 or RPGRIP1L KD cells. Our results demonstrate that the RPGR protein complex is required for regulating proteasomal activity and for modulating SOCE, which may contribute to the ciliopathy phenotype.

14.
Food Chem Toxicol ; 112: 76-85, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29274434

ABSTRACT

Oxidative stress plays a critical role in the pathogenesis of retinal degeneration. Gypenosides are the major functional components isolated from Gynostemma pentaphyllum. They have been shown to protect against oxidative stress and inflammation and have also demonstrated a protective effect on experimental optic neuritis. In order to determine the protective properties of gypenosides against oxidative stress in human retinal pigment epithelium (RPE) cells, ARPE-19 cells were treated with H2O2 or H2O2 plus gypenosides for 24 h. ARPE-19 cells co-treated with gypenosides had significantly increased cell viability and decreased cell death rate when compared to cells treated with H2O2 alone. The level of GSH, the activities of SOD and catalase, and the expression of NRF2 and antioxidant genes were notably decreased, while there were marked increases in ROS, MDA and pro-inflammatory cytokines in ARPE-19 cells exposed to H2O2; co-treatment with gypenosides significantly counteract these changes. Our study suggests that gypenosides protect RPE cells from oxidative damage and offer therapeutic potential for the treatment of retinal degeneration.


Subject(s)
Oxidative Stress/drug effects , Retinal Pigment Epithelium/drug effects , Antioxidants/metabolism , Catalase/metabolism , Cell Death/drug effects , Cell Line , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/drug effects , Glutathione/metabolism , Gynostemma , Humans , Hydrogen Peroxide/pharmacology , In Situ Nick-End Labeling , Inflammation/genetics , Malondialdehyde/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Retinal Degeneration/drug therapy , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Signal Transduction , Superoxide Dismutase/metabolism , Up-Regulation/drug effects
15.
Sci Rep ; 7(1): 16881, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203866

ABSTRACT

Mutations in the RPGR-interacting protein 1 (RPGRIP1) gene cause recessive Leber congenital amaurosis (LCA), juvenile retinitis pigmentosa (RP) and cone-rod dystrophy. RPGRIP1 interacts with other retinal disease-causing proteins and has been proposed to have a role in ciliary protein transport; however, its function remains elusive. Here, we describe a new zebrafish model carrying a nonsense mutation in the rpgrip1 gene. Rpgrip1homozygous mutants do not form rod outer segments and display mislocalization of rhodopsin, suggesting a role for RPGRIP1 in rhodopsin-bearing vesicle trafficking. Furthermore, Rab8, the key regulator of rhodopsin ciliary trafficking, was mislocalized in photoreceptor cells of rpgrip1 mutants. The degeneration of rod cells is early onset, followed by the death of cone cells. These phenotypes are similar to that observed in LCA and juvenile RP patients. Our data indicate RPGRIP1 is necessary for rod outer segment development through regulating ciliary protein trafficking. The rpgrip1 mutant zebrafish may provide a platform for developing therapeutic treatments for RP patients.


Subject(s)
Cilia/metabolism , Rod Cell Outer Segment/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Codon, Nonsense , Protein Transport , Retina/metabolism , Retina/pathology , Retinal Degeneration/pathology , Rhodopsin/metabolism , Zebrafish/growth & development , Zebrafish Proteins/genetics , rab GTP-Binding Proteins/metabolism
16.
Hum Mol Genet ; 26(22): 4327-4339, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28973423

ABSTRACT

Cholesterol accumulation beneath the retinal pigment epithelium (RPE) cells is supposed to contribute the pathogenesis of age-related macular degeneration (AMD). Cholesterol efflux genes (APOE and ABCA1) were identified as risk factors for AMD, although how cholesterol efflux influences accumulation of this lipid in sub-RPE deposits remains elusive. The 18 kDa translocator protein, TSPO, is a cholesterol-binding protein implicated in mitochondrial cholesterol transport. Here, we investigate the function of TSPO in cholesterol efflux from the RPE cells. We demonstrate in RPE cells that TSPO specific ligands promoted cholesterol efflux to acceptor (apo)lipoprotein and human serum, while loss of TSPO resulted in impaired cholesterol efflux. TSPO-/- RPE cells also had significantly increased production of reactive oxygen species (ROS) and upregulated expression of proinflammatory cytokines (IL-1ß and TNFα). Cholesterol (oxidized LDL) uptake and accumulation were markedly increased in TSPO-/- RPE cells. Finally, in aged RPE cells, TSPO expression was reduced and cholesterol efflux impaired. These findings provide a new pharmacological concept to treat early AMD patients by stimulating cellular cholesterol removal with TSPO specific ligands or by overexpression of TSPO in RPE cells.


Subject(s)
Cholesterol/metabolism , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Receptors, GABA/metabolism , Retinal Pigment Epithelium/metabolism , Biological Transport , Carrier Proteins/metabolism , Cells, Cultured , Humans , Indoleacetic Acids/pharmacology , Ligands , Lipoproteins, LDL/metabolism , Mitochondria/metabolism , Molecular Targeted Therapy , Oxazines/pharmacology , Oxidative Stress , Pancreatic Elastase/metabolism , Purines/pharmacology , Reactive Oxygen Species/metabolism
17.
Hum Mol Genet ; 26(12): 2335-2345, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28398482

ABSTRACT

In humans, CERKL mutations cause widespread retinal degeneration: early dysfunction and loss of rod and cone photoreceptors in the outer retina and, progressively, death of cells in the inner retina. Despite intensive efforts, the function of CERKL remains obscure and studies in animal models have failed to clarify the disease mechanism of CERKL mutations. To address this gap in knowledge, we have generated a stable CERKL knockout zebrafish model by TALEN technology and a 7bp deletion in CERKL cDNA that caused the premature termination of CERKL. These CERKL-/- animals showed progressive degeneration of photoreceptor outer segments (OSs) and increased apoptosis of retinal cells, including those in the outer and inner retinal layers. Additionally, we confirmed by immunofluorescence and western-blot that rod degeneration in CERKL-/- zebrafish occurred earlier and was more significant than that in cone cells. Accumulation of shed OSs in the interphotoreceptor matrix was observed by transmission election microscopy (TEM). This suggested that CERKL may regulate the phagocytosis of OSs by the retinal pigment epithelium (RPE). We further found that the phagocytosis-associated protein MERTK was significantly reduced in CERKL-/- zebrafish. Additionally, in ARPE-19 cell lines, knockdown of CERKL also decreased the mRNA and protein level of MERTK, as well as the ox-POS phagocytosis. We conclude that CERKL deficiency in zebrafish may cause rod-cone dystrophy, but not cone-rod dystrophy, while interfering with the phagocytosis function of RPE associated with down-regulation of the expression of MERTK.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Animals , Cell Line , Down-Regulation , Gene Knockout Techniques/methods , Humans , Mutation , Phagocytosis/genetics , Photoreceptor Cells , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Retinal Pigment Epithelium/metabolism , Retinitis Pigmentosa/metabolism , Zebrafish/genetics
18.
Food Chem Toxicol ; 108(Pt B): 543-553, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28159594

ABSTRACT

Acrylamide is a substance that can be neurotoxic in humans and experimental animals. It is formed at different rates in starchy foods cooked at temperatures above 120 °C as a result of interaction between monosaccharides and the amino acid asparagine. Carnosic acid accounts for over 90% of the antioxidant properties of rosemary extract and is a powerful inhibitor of lipid peroxidation in microsomal and liposomal systems. Carnosic acid has been shown to protect against oxidative and inflammatory effects. In order to investigate the protective properties of carnosic acid against acrylamide-induced toxicity in human retinal pigment epithelium (RPE) cells, ARPE-19 cells were pre-treated with 10 µM carnosic acid for 24 h followed by treatment with acrylamide (0.7 or 1 mM) for 24 h. ARPE-19 cells pre-treated with 10 µM carnosic acid showed significantly increased cell viability and decreased cell death rate when compared to ARPE-19 cells treated with acrylamide alone. Activities of SOD and catalase and the level of GSH and expression of NRF2 and a number of anti-oxidant genes were significantly decreased in ARPE-19 cells, while there were significant increases in ROS and MDA; pre-treatment with carnosic acid significantly counteracted these changes. Our results suggest that carnosic acid protected RPE cells from acrylamide-induced toxicity.


Subject(s)
Abietanes/pharmacology , Acrylamide/toxicity , Epithelial Cells/drug effects , Retinal Pigment Epithelium/cytology , Abietanes/chemistry , Acrylamide/chemistry , Cell Line , Gene Expression Regulation/drug effects , Humans , Molecular Structure , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL