124 results on '"Antonetti DA"'
Search Results
2. Protein kinase cβ phosphorylates occludin regulating tight junction trafficking in vascular endothelial growth factor-induced permeability in vivo.
- Author
-
Murakami T, Frey T, Lin C, Antonetti DA, Murakami, Tomoaki, Frey, Tiffany, Lin, Chengmao, and Antonetti, David A
- Abstract
Vascular endothelial growth factor (VEGF)-induced breakdown of the blood-retinal barrier requires protein kinase C (PKC)β activation. However, the molecular mechanisms related to this process remain poorly understood. In this study, the role of occludin phosphorylation and ubiquitination downstream of PKCβ activation in tight junction (TJ) trafficking and endothelial permeability was investigated. Treatment of bovine retinal endothelial cells and intravitreal injection of PKCβ inhibitors as well as expression of dominant-negative kinase was used to determine the contribution of PKCβ to endothelial permeability and occludin phosphorylation at Ser490 detected with a site-specific antibody. In vitro kinase assay was used to demonstrate direct occludin phosphorylation by PKCβ. Ubiquitination was measured by immunoblotting after occludin immunoprecipitation. Confocal microscopy revealed organization of TJ proteins. The results reveal that inhibition of VEGF-induced PKCβ activation blocks occludin Ser490 phosphorylation, ubiquitination, and TJ trafficking in retinal vascular endothelial cells both in vitro and in vivo and prevents VEGF-stimulated vascular permeability. Occludin Ser490 is a direct target of PKCβ, and mutating Ser490 to Ala (S490A) blocks permeability downstream of PKCβ. Therefore, PKCβ activation phosphorylates occludin on Ser490, leading to ubiquitination required for VEGF-induced permeability. These data demonstrate a novel mechanism for PKCβ targeted inhibitors in regulating vascular permeability. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
3. TNF-α signals through PKCζ/NF-κB to alter the tight junction complex and increase retinal endothelial cell permeability.
- Author
-
Aveleira CA, Lin CM, Abcouwer SF, Ambrósio AF, Antonetti DA, Aveleira, Célia A, Lin, Cheng-Mao, Abcouwer, Steven F, Ambrósio, António F, and Antonetti, David A
- Abstract
Objective: Tumor necrosis factor-α (TNF-α) and interleukin-1 beta (IL-1β) are elevated in the vitreous of diabetic patients and in retinas of diabetic rats associated with increased retinal vascular permeability. However, the molecular mechanisms underlying retinal vascular permeability induced by these cytokines are poorly understood. In this study, the effects of IL-1β and TNF-α on retinal endothelial cell permeability were compared and the molecular mechanisms by which TNF-α increases cell permeability were elucidated.Research Design and Methods: Cytokine-induced retinal vascular permeability was measured in bovine retinal endothelial cells (BRECs) and rat retinas. Western blotting, quantitative real-time PCR, and immunocytochemistry were performed to determine tight junction protein expression and localization.Results: IL-1β and TNF-α increased BREC permeability, and TNF-α was more potent. TNF-α decreased the protein and mRNA content of the tight junction proteins ZO-1 and claudin-5 and altered the cellular localization of these tight junction proteins. Dexamethasone prevented TNF-α-induced cell permeability through glucocorticoid receptor transactivation and nuclear factor-kappaB (NF-κB) transrepression. Preventing NF-κB activation with an inhibitor κB kinase (IKK) chemical inhibitor or adenoviral overexpression of inhibitor κB alpha (IκBα) reduced TNF-α-stimulated permeability. Finally, inhibiting protein kinase C zeta (PKCζ) using both a peptide and a novel chemical inhibitor reduced NF-κB activation and completely prevented the alterations in the tight junction complex and cell permeability induced by TNF-α in cell culture and rat retinas.Conclusions: These results suggest that PKCζ may provide a specific therapeutic target for the prevention of vascular permeability in retinal diseases characterized by elevated TNF-α, including diabetic retinopathy. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
4. Ablation of 4E-BP1/2 prevents hyperglycemia-mediated induction of VEGF expression in the rodent retina and in Muller cells in culture.
- Author
-
Schrufer TL, Antonetti DA, Sonenberg N, Kimball SR, Gardner TW, Jefferson LS, Schrufer, Tabitha L, Antonetti, David A, Sonenberg, Nahum, Kimball, Scot R, Gardner, Thomas W, and Jefferson, Leonard S
- Abstract
Objective: Vascular endothelial growth factor (VEGF) contributes to diabetic retinopathy, but control of its expression is not well understood. Here, we tested the hypothesis that hyperglycemia mediates induction of VEGF expression in a eukaryotic initiation factor 4E (eIF4E) binding protein (4E-BP) 1 and 2 dependent manner.Research Design and Methods: The retina was harvested from control and type 1 diabetic rats and mice and analyzed for VEGF mRNA and protein expression as well as biomarkers of translational control mechanisms. Similar analyses were performed in Müller cell cultures exposed to hyperglycemic conditions. The effect of 4E-BP1 and 4E-BP2 gene deletion on VEGF expression was examined in mice and in mouse embryo fibroblasts (MEFs).Results: Whereas VEGF mRNA in the retina remained constant, VEGF expression was increased as early as 2 weeks after the onset of diabetes. Increases in expression of 4E-BP1 protein mirrored those of VEGF and expression of 4E-BP1 mRNA was unchanged. Similar results were observed after 10 h of exposure of cells in culture to hyperglycemic conditions. Importantly, the diabetes-induced increase in VEGF expression was not observed in mice deficient in 4E-BP1 and 4E-BP2, nor in MEFs lacking the two proteins.Conclusions: Hyperglycemia induces VEGF expression through cap-independent mRNA translation mediated by increased expression of 4E-BP1. Because the VEGF mRNA contains two internal ribosome entry sites, the increased expression is likely a consequence of ribosome loading at these sites. These findings provide new insights into potential targets for treatment of diabetic retinopathy. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
5. Diabetes reduces basal retinal insulin receptor signaling: reversal with systemic and local insulin.
- Author
-
Reiter CEN, Wu X, Sandirasegarane L, Nakamura M, Gilbert KA, Singh RSF, Fort PE, Antonetti DA, Gardner TW, Reiter, Chad E N, Wu, Xiaohua, Sandirasegarane, Lakshman, Nakamura, Makoto, Gilbert, Kirk A, Singh, Ravi S J, Fort, Patrice E, Antonetti, David A, and Gardner, Thomas W
- Abstract
Diabetic retinopathy is characterized by early onset of neuronal cell death. We previously showed that insulin mediates a prosurvival pathway in retinal neurons and that normal retina expresses a highly active basal insulin receptor/Akt signaling pathway that is stable throughout feeding and fasting. Using the streptozotocin-induced diabetic rat model, we tested the hypothesis that diabetes diminishes basal retinal insulin receptor signaling concomitantly with increased diabetes-induced retinal apoptosis. The expression, phosphorylation status, and/or kinase activity of the insulin receptor and downstream signaling proteins were investigated in retinas of age-matched control, diabetic, and insulin-treated diabetic rats. Four weeks of diabetes reduced basal insulin receptor kinase, insulin receptor substrate (IRS)-1/2-associated phosphatidylinositol 3-kinase, and Akt kinase activity without altering insulin receptor or IRS-1/2 expression or tyrosine phosphorylation. After 12 weeks of diabetes, constitutive insulin receptor autophosphorylation and IRS-2 expression were reduced, without changes in p42/p44 mitogen-activated protein kinase or IRS-1. Sustained systemic insulin treatment of diabetic rats prevented loss of insulin receptor and Akt kinase activity, and acute intravitreal insulin administration restored insulin receptor kinase activity. Insulin treatment restored insulin receptor-beta autophosphorylation in rat retinas maintained ex vivo, demonstrating functional receptors and suggesting loss of ligand as a cause for reduced retinal insulin receptor/Akt pathway activity. These results demonstrate that diabetes progressively impairs the constitutive retinal insulin receptor signaling pathway through Akt and suggests that loss of this survival pathway may contribute to the initial stages of diabetic retinopathy. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
6. Whole genome assessment of the retinal response to diabetes reveals a progressive neurovascular inflammatory response
- Author
-
Brucklacher Robert M, Patel Kruti M, VanGuilder Heather D, Bixler Georgina V, Barber Alistair J, Antonetti David A, Lin Cheng-Mao, LaNoue Kathryn F, Gardner Thomas W, Bronson Sarah K, and Freeman Willard M
- Subjects
Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background Despite advances in the understanding of diabetic retinopathy, the nature and time course of molecular changes in the retina with diabetes are incompletely described. This study characterized the functional and molecular phenotype of the retina with increasing durations of diabetes. Results Using the streptozotocin-induced rat model of diabetes, levels of retinal permeability, caspase activity, and gene expression were examined after 1 and 3 months of diabetes. Gene expression changes were identified by whole genome microarray and confirmed by qPCR in the same set of animals as used in the microarray analyses and subsequently validated in independent sets of animals. Increased levels of vascular permeability and caspase-3 activity were observed at 3 months of diabetes, but not 1 month. Significantly more and larger magnitude gene expression changes were observed after 3 months than after 1 month of diabetes. Quantitative PCR validation of selected genes related to inflammation, microvasculature and neuronal function confirmed gene expression changes in multiple independent sets of animals. Conclusion These changes in permeability, apoptosis, and gene expression provide further evidence of progressive retinal malfunction with increasing duration of diabetes. The specific gene expression changes confirmed in multiple sets of animals indicate that pro-inflammatory, anti-vascular barrier, and neurodegenerative changes occur in tandem with functional increases in apoptosis and vascular permeability. These responses are shared with the clinically documented inflammatory response in diabetic retinopathy suggesting that this model may be used to test anti-inflammatory therapeutics.
- Published
- 2008
- Full Text
- View/download PDF
7. Publisher Correction: Current understanding of the molecular and cellular pathology of diabetic retinopathy.
- Author
-
Antonetti DA, Silva PS, and Stitt AW
- Published
- 2025
- Full Text
- View/download PDF
8. Diabetes Renders Photoreceptors Susceptible to Retinal Ischemia-Reperfusion Injury.
- Author
-
Antonetti DA, Lin CM, Shanmugam S, Hager H, Cao M, Liu X, Dreffs A, Habash A, and Abcouwer SF
- Subjects
- Animals, Mice, Photoreceptor Cells, Vertebrate pathology, Photoreceptor Cells, Vertebrate drug effects, Male, Phlorhizin pharmacology, Capillary Permeability, Diabetes Mellitus, Type 2 complications, Apoptosis, Reperfusion Injury metabolism, Mice, Inbred C57BL, Diabetes Mellitus, Experimental complications, Diabetic Retinopathy etiology, Diabetic Retinopathy metabolism, In Situ Nick-End Labeling
- Abstract
Purpose: Studies have suggested that photoreceptors (PR) are altered by diabetes, contributing to diabetic retinopathy (DR) pathology. Here, we explored the effect of diabetes on retinal ischemic injury., Methods: Retinal ischemia-reperfusion (IR) injury was caused by elevation of intraocular pressure in 10-week-old BKS db/db type 2 diabetes mellitus (T2DM) mice or C57BL/6J mice at 4 or 12 weeks after streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM), and respective nondiabetic controls. Retinal neurodegeneration was evaluated by retinal layer thinning, TUNEL staining, and neuron loss. Vascular permeability was evaluated as retinal accumulation of circulating fluorescent albumin. The effects of pretreatment with a sodium-glucose co-transporter (SGLT1/2) inhibitor, phlorizin, were examined., Results: Nondiabetic control mice exhibited no significant outer retinal layer thinning or PR loss after IR injury. In contrast, db/db mice exhibited significant outer retina thinning (49%, P < 0.0001), loss of PR nuclei (45%, P < 0.05) and inner segment (IS) length decline (45%, P < 0.0001). STZ-induced diabetic mice at 4 weeks showed progressive thinning of the outer retina (55%, by 14 days, P < 0.0001) and 4.3-fold greater number of TUNEL+ cells in the outer nuclear layer (ONL) than injured retinas of control mice (P < 0.0001). After 12 weeks of diabetes, the retinas exhibited similar outer layer thinning and PR loss after IR. Diabetes also delayed restoration of the blood-retinal barrier after IR injury. Phlorizin reduced outer retinal layer thinning from 49% to 3% (P < 0.0001)., Conclusions: Diabetes caused PR to become highly susceptible to IR injury. The ability of phlorizin pretreatment to block outer retinal thinning after IR suggests that the effects of diabetes on PR are readily reversible.
- Published
- 2024
- Full Text
- View/download PDF
9. Imaging Modalities for Assessing the Vascular Component of Diabetic Retinal Disease: Review and Consensus for an Updated Staging System.
- Author
-
Tan TE, Jampol LM, Ferris FL, Tadayoni R, Sadda SR, Chong V, Domalpally A, Blodi BL, Duh EJ, Curcio CA, Antonetti DA, Dutta S, Levine SR, Sun JK, Gardner TW, and Wong TY
- Abstract
Purpose: To review the evidence for imaging modalities in assessing the vascular component of diabetic retinal disease (DRD), to inform updates to the DRD staging system., Design: Standardized narrative review of the literature by an international expert workgroup, as part of the DRD Staging System Update Effort, a project of the Mary Tyler Moore Vision Initiative. Overall, there were 6 workgroups: Vascular Retina, Neural Retina, Systemic Health, Basic and Cellular Mechanisms, Visual Function, and Quality of Life., Participants: The Vascular Retina workgroup, including 16 participants from 4 countries., Methods: Literature review was conducted using standardized evidence grids for 5 modalities: standard color fundus photography (CFP), widefield color photography (WFCP), standard fluorescein angiography (FA), widefield FA (WFFA), and OCT angiography (OCTA). Summary levels of evidence were determined on a validated scale from I (highest) to V (lowest). Five virtual workshops were held for discussion and consensus., Main Outcome Measures: Level of evidence for each modality., Results: Levels of evidence for standard CFP, WFCP, standard FA, WFFA, and OCTA were I, II, I, I, and II respectively. Traditional vascular lesions on standard CFP should continue to be included in an updated staging system, but more studies are required before they can be used in posttreatment eyes. Widefield color photographs can be used for severity grading within the area covered by standard CFPs, although these gradings may not be directly interchangeable with each other. Evaluation of the peripheral retina on WFCP can be considered, but the method of grading needs to be clarified and validated. Standard FA and WFFA provide independent prognostic value, but the need for dye administration should be considered. OCT angiography has significant potential for inclusion in the DRD staging system, but various barriers need to be addressed first., Conclusions: This study provides evidence-based recommendations on the utility of various imaging modalities for assessment of the vascular component of DRD, which can inform future updates to the DRD staging system. Although new imaging modalities offer a wealth of information, there are still major gaps and unmet research needs that need to be addressed before this potential can be realized., Financial Disclosures: Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article., (© 2023 by the American Academy of Ophthalmology.)
- Published
- 2023
- Full Text
- View/download PDF
10. Disheveled-1 Interacts with Claudin-5 and Contributes to Norrin-Induced Endothelial Barrier Restoration.
- Author
-
Díaz-Coránguez M, González-González L, Wang A, Liu X, and Antonetti DA
- Subjects
- Rats, Humans, Animals, Cattle, Claudin-5 genetics, Vascular Endothelial Growth Factor A metabolism, HEK293 Cells, beta Catenin metabolism, Endothelial Cells metabolism
- Abstract
Previous studies have revealed that norrin can reverse vascular endothelial-growth-factor (VEGF)-induced permeability in a β-catenin-dependent pathway. Here, we have explored the contribution of disheveled-1 (DVL1) in norrin-induced blood-retinal barrier (BRB) restoration. We provide evidence that in addition to canonical signaling, DVL1 promotes tight junction (TJ) stabilization through a novel, non-canonical signaling pathway involving direct claudin-5 (CLDN5) binding. Immunofluorescence staining of rat retinal cross-sections showed enriched expression of DVL1 and 3 at endothelial capillaries and co-localization with CLDN5 and ZO-1 at the TJ complex in primary bovine retinal endothelial cells (BRECs). Barrier properties of BRECs were determined via measurements of trans-endothelial electrical resistance (TEER) or permeability to 70 kDa RITC-dextran. These studies demonstrated that norrin restoration of barrier properties after VEGF treatment required DVL1 as an siRNA knockdown of Dvl1 but not Dvl2 or Dvl3, reduced basal barrier properties and ablated norrin-induced barrier restoration. However, loss of Dvl1 did not decrease β-catenin signaling activity as measured by Axin2 mRNA expression, suggesting the contribution of a non-canonical pathway. DVL and TJ protein interactions were analyzed via co-immunoprecipitation of endogenous protein in BRECs, which demonstrated that DVL1 interacts with both CLDN5 and ZO-1, while DVL3 interacts only with ZO-1. These interactions were most abundant after inducing BRB restoration by treating BRECs with VEGF and norrin. DVL has previously been shown to form intramolecular bindings between the C-terminal PDZ-binding motif (PDZ-BM) with an internal PDZ domain. Co-transfection of HEK293 cells with DVL1 and CLDN5 or relevant mutants revealed that DVL1 interacts with CLDN5 through the DVL PDZ domain binding, CLDN5 PDZ-BM, in competition with DVL1 PDZ-BM, since DVL/CLDN5 interaction increases with deletion of the DVL1 PDZ-BM and decreases by co-expressing the C-terminal fragment of DVL1 containing the PDZ-BM or through deletion of CLDN5 PDZ-BM. In BREC cells, transfection of the C-terminal fragment of DVL1 downregulates the expression of CLDN5 but does not affect the expression of other proteins of the TJs, including ZO-1, occludin, CLDN1 or VE-cadherin. Blocking DVL1/CLDN5 interaction increased basal permeability and prevented norrin induction of barrier properties after VEGF. Combined with previous data, these results demonstrate that norrin signals through both a canonical β-catenin pathway and a non-canonical signaling pathway by which DVL1 directly binds to CLDN5 to promote barrier properties.
- Published
- 2023
- Full Text
- View/download PDF
11. Delineating effects of angiopoietin-2 inhibition on vascular permeability and inflammation in models of retinal neovascularization and ischemia/reperfusion.
- Author
-
Canonica J, Foxton R, Garrido MG, Lin CM, Uhles S, Shanmugam S, Antonetti DA, Abcouwer SF, and Westenskow PD
- Abstract
Introduction: Clinical trials demonstrated that co-targeting angiopoietin-2 (Ang-2) and vascular endothelial growth factor (VEGF-A) with faricimab controls anatomic outcomes and maintains vision improvements, with strong durability, through 2 years in patients with neovascular age-related macular degeneration and diabetic macular edema. The mechanism(s) underlying these findings is incompletely understood and the specific role that Ang-2 inhibition plays requires further investigation., Methods: We examined the effects of single and dual Ang-2/VEGF-A inhibition in diseased vasculatures of JR5558 mice with spontaneous choroidal neovascularization (CNV) and in mice with retinal ischemia/reperfusion (I/R) injuries., Results: In JR5558 mice, Ang-2, VEGF-A, and dual Ang-2/VEGF-A inhibition reduced CNV area after 1 week; only dual Ang-2/VEGF-A inhibition decreased neovascular leakage. Only Ang-2 and dual Ang-2/VEGF-A inhibition maintained reductions after 5 weeks. Dual Ang-2/VEGF-A inhibition reduced macrophage/microglia accumulation around lesions after 1 week. Both Ang-2 and dual Ang-2/VEGF-A inhibition reduced macrophage/microglia accumulation around lesions after 5 weeks. In the retinal I/R injury model, dual Ang-2/VEGF-A inhibition was statistically significantly more effective than Ang-2 or VEGF-A inhibition alone in preventing retinal vascular leakage and neurodegeneration., Discussion: These data highlight the role of Ang-2 in dual Ang-2/VEGF-A inhibition and indicate that dual inhibition has complementary anti-inflammatory and neuroprotective effects, suggesting a mechanism for the durability and efficacy of faricimab in clinical trials., Competing Interests: JC, RF, SU, and PW are employees of F. Hoffmann-La Roche Ltd., Roche Pharma AG, or Genentech, Inc. MG was an employee of F. Hoffmann-La Roche Ltd. at the time the studies described in this manuscript were performed and is a current employee of Boehringer Ingelheim (Schweiz) GmbH. DA and SA have received research funding from F. Hoffmann-La Roche Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. This study received funding from F. Hoffmann-La Roche Ltd. The funder had the following involvement with the study: study design; conducting the study; data collection, management, analysis, interpretation, preparation, review, and approval of the manuscript. Funding was provided by F. Hoffmann-La Roche Ltd. for third-party writing assistance, which was provided by Luke Carey, Ph.D., CMPP, of Envision Pharma Group., (Copyright © 2023 Canonica, Foxton, Garrido, Lin, Uhles, Shanmugam, Antonetti, Abcouwer and Westenskow.)
- Published
- 2023
- Full Text
- View/download PDF
12. Transgenic animal models to explore and modulate the blood brain and blood retinal barriers of the CNS.
- Author
-
Goncalves A and Antonetti DA
- Subjects
- Animals, Animals, Genetically Modified, Endothelial Cells physiology, Central Nervous System, Blood-Retinal Barrier metabolism, Blood-Brain Barrier metabolism
- Abstract
The unique environment of the brain and retina is tightly regulated by blood-brain barrier and the blood-retinal barrier, respectively, to ensure proper neuronal function. Endothelial cells within these tissues possess distinct properties that allow for controlled passage of solutes and fluids. Pericytes, glia cells and neurons signal to endothelial cells (ECs) to form and maintain the barriers and control blood flow, helping to create the neurovascular unit. This barrier is lost in a wide range of diseases affecting the central nervous system (CNS) and retina such as brain tumors, stroke, dementia, and in the eye, diabetic retinopathy, retinal vein occlusions and age-related macular degeneration to name prominent examples. Recent studies directly link barrier changes to promotion of disease pathology and degradation of neuronal function. Understanding how these barriers form and how to restore these barriers in disease provides an important point for therapeutic intervention. This review aims to describe the fundamentals of the blood-tissue barriers of the CNS and how the use of transgenic animal models led to our current understanding of the molecular framework of these barriers. The review also highlights examples of targeting barrier properties to protect neuronal function in disease states., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
13. Thrombolytic tPA-induced hemorrhagic transformation of ischemic stroke is mediated by PKCβ phosphorylation of occludin.
- Author
-
Goncalves A, Su EJ, Muthusamy A, Zeitelhofer M, Torrente D, Nilsson I, Protzmann J, Fredriksson L, Eriksson U, Antonetti DA, and Lawrence DA
- Subjects
- Animals, Cerebral Hemorrhage drug therapy, Cerebral Hemorrhage etiology, Fibrinolytic Agents therapeutic use, Humans, Infarction, Middle Cerebral Artery drug therapy, Mice, Occludin genetics, Occludin metabolism, Phosphorylation, Thrombolytic Therapy adverse effects, Thrombolytic Therapy methods, Tissue Plasminogen Activator metabolism, Ischemic Stroke, Stroke complications, Stroke etiology
- Abstract
The current standard of care for moderate to severe ischemic stroke is thrombolytic therapy with tissue plasminogen activator (tPA). Treatment with tPA can significantly improve neurologic outcomes; however, thrombolytic therapy is associated with an increased risk of intracerebral hemorrhage (ICH). The risk of hemorrhage significantly limits the use of thrombolytic therapy, and identifying pathways induced by tPA that increase this risk could provide new therapeutic options to extend thrombolytic therapy to a wider patient population. Here, we investigate the role of protein kinase Cβ (PKCβ) phosphorylation of the tight junction protein occludin during ischemic stroke and its role in cerebrovascular permeability. We show that activation of this pathway by tPA is associated with an increased risk of ICH. Middle cerebral artery occlusion (MCAO) increased phosphorylation of occludin serine 490 (S490) in the ischemic penumbra in a tPA-dependent manner, as tPA-/- mice were significantly protected from MCAO-induced occludin phosphorylation. Intraventricular injection of tPA in the absence of ischemia was sufficient to induce occludin phosphorylation and vascular permeability in a PKCβ-dependent manner. Blocking occludin phosphorylation, either by targeted expression of a non-phosphorylatable form of occludin (S490A) or by pharmacologic inhibition of PKCβ, reduced MCAO-induced permeability and improved functional outcome. Furthermore, inhibiting PKCβ after MCAO prevented ICH associated with delayed thrombolysis. These results show that PKCβ phosphorylation of occludin is a downstream mediator of tPA-induced cerebrovascular permeability and suggest that PKCβ inhibitors could improve stroke outcome and prevent ICH associated with delayed thrombolysis, potentially extending the window for thrombolytic therapy in stroke., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
14. Structure and function of the retina of low-density lipoprotein receptor-related protein 5 (Lrp5)-deficient rats.
- Author
-
Ubels JL, Lin CM, Antonetti DA, Diaz-Coranguez M, Diegel CR, and Williams BO
- Subjects
- Animals, Claudin-5 biosynthesis, Claudin-5 genetics, Evans Blue pharmacology, Familial Exudative Vitreoretinopathies genetics, Familial Exudative Vitreoretinopathies metabolism, Mutation, Rats, Structure-Activity Relationship, Low Density Lipoprotein Receptor-Related Protein-5 genetics, Low Density Lipoprotein Receptor-Related Protein-5 metabolism, Retina metabolism
- Abstract
Loss-of-function mutations in the Wnt co-receptor, low-density lipoprotein receptor-related protein 5 (LRP5), result in familial exudative vitreoretinopathy (FEVR), osteoporosis-pseudoglioma syndrome (OPPG), and Norrie disease. CRISPR/Cas9 gene editing was used to produce rat strains deficient in Lrp5. The purpose of this study was to validate this rat model for studies of hypovascular, exudative retinopathies. The retinal vasculature of wildtype and Lrp5 knockout rats was stained with Giffonia simplifolia isolectin B
4 and imaged by fluorescence microscopy. Effects on retinal structure were investigated by histology. The integrity of the blood-retina barrier was analyzed by measurement of permeability to Evans blue dye and staining for claudin-5. Retinas were imaged by fundus photography and SD-OCT, and electroretinograms were recorded. Lrp5 gene deletion led to sparse superficial retinal capillaries and loss of the deep and intermediate plexuses. Autofluorescent exudates were observed and are correlated with increased Evans blue permeability and absence of claudin-5 expression in superficial vessels. OCT images show pathology similar to OCT of humans with FEVR, and retinal thickness is reduced by 50% compared to wild-type rats. Histology and OCT reveal that photoreceptor and outer plexiform layers are absent. The retina failed to demonstrate an ERG response. CRISPR/Cas9 gene-editing produced a predictable rat Lrp5 knockout model with extensive defects in the retinal vascular and neural structure and function. This rat model should be useful for studies of exudative retinal vascular diseases involving the Wnt and norrin pathways., (Copyright © 2022 Elsevier Ltd. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
15. Regulation of Adrenergic, Serotonin, and Dopamine Receptors to Inhibit Diabetic Retinopathy: Monotherapies versus Combination Therapies.
- Author
-
Kern TS, Du Y, Tang J, Lee CA, Liu H, Dreffs A, Leinonen H, Antonetti DA, and Palczewski K
- Subjects
- Animals, Diabetes Mellitus, Experimental drug therapy, Diabetes Mellitus, Experimental metabolism, Diabetic Retinopathy pathology, Dose-Response Relationship, Drug, Drug Therapy, Combination, Male, Mice, Mice, Inbred C57BL, Retinal Vessels drug effects, Retinal Vessels metabolism, Retinal Vessels pathology, Diabetic Retinopathy drug therapy, Diabetic Retinopathy metabolism, Hypoglycemic Agents administration & dosage, Receptors, Adrenergic metabolism, Receptors, Dopamine metabolism, Receptors, Serotonin metabolism
- Abstract
We compared monotherapies and combinations of therapies that regulate G-protein-coupled receptors (GPCRs) with respect to their abilities to inhibit early stages of diabetic retinopathy (DR) in streptozotocin-diabetic mice. Metoprolol (MTP; 0.04-1.0 mg/kg b.wt./day), bromocriptine (BRM; 0.01-0.1 mg/kg b.wt./day), doxazosin (DOX; 0.01-1.0 mg/kg b.wt./day), or tamsulosin (TAM; 0.05-0.25 mg/kg b.wt./day) were injected individually daily for 2 months in dose-response studies to assess their effects on the diabetes-induced increases in retinal superoxide and leukocyte-mediated cytotoxicity against vascular endothelial cells, both of which abnormalities have been implicated in the development of DR. Each of the individual drugs inhibited the diabetes-induced increase in retinal superoxide at the higher concentrations tested, but the inhibition was lost at lower doses. To determine whether combination therapies had superior effects over individual drugs, we intentionally selected for each drug a low dose that had little or no effect on the diabetes-induced retinal superoxide for use separately or in combinations in 8-month studies of retinal function, vascular permeability, and capillary degeneration in diabetes. At the low doses used, combinations of the drugs generally were more effective than individual drugs, but the low-dose MTP alone totally inhibited diabetes-induced reduction in a vision task, BRM or DOX alone totally inhibited the vascular permeability defect, and DOX alone totally inhibited diabetes-induced degeneration of retinal capillaries. Although low-dose MTP, BRM, DOX, or TAM individually had beneficial effects on some endpoints, combination of the therapies better inhibited the spectrum of DR lesions evaluated. SIGNIFICANCE STATEMENT: The pathogenesis of early stages of diabetic retinopathy remains incompletely understood, but multiple different cell types are believed to be involved in the pathogenic process. We have compared the effects of monotherapies to those of combinations of drugs that regulate GPCR signaling pathways with respect to their relative abilities to inhibit the development of early diabetic retinopathy., (Copyright © 2021 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2021
- Full Text
- View/download PDF
16. Inflammatory resolution and vascular barrier restoration after retinal ischemia reperfusion injury.
- Author
-
Abcouwer SF, Shanmugam S, Muthusamy A, Lin CM, Kong D, Hager H, Liu X, and Antonetti DA
- Subjects
- Animals, Apoptosis physiology, Capillary Permeability physiology, DNA Fragmentation, Disease Models, Animal, Mice, Microglia metabolism, Recovery of Function physiology, Blood-Retinal Barrier pathology, Inflammation pathology, Reperfusion Injury pathology, Retina pathology, Retinal Vessels pathology
- Abstract
Background: Several retinal pathologies exhibit both inflammation and breakdown of the inner blood-retinal barrier (iBRB) resulting in vascular permeability, suggesting that treatments that trigger resolution of inflammation may also promote iBRB restoration., Methods: Using the mouse retinal ischemia-reperfusion (IR) injury model, we followed the time course of neurodegeneration, inflammation, and iBRB disruption and repair to examine the relationship between resolution of inflammation and iBRB restoration and to determine if minocycline, a tetracycline derivative shown to reverse microglial activation, can hasten these processes., Results: A 90-min ischemic insult followed by reperfusion in the retina induced cell apoptosis and inner retina thinning that progressed for approximately 2 weeks. IR increased vascular permeability within hours, which resolved between 3 and 4 weeks after injury. Increased vascular permeability coincided with alteration and loss of endothelial cell tight junction (TJ) protein content and disorganization of TJ protein complexes. Shunting of blood flow away from leaky vessels and dropout of leaky capillaries were eliminated as possible mechanisms for restoring the iBRB. Repletion of TJ protein contents occurred within 2 days after injury, long before restoration of the iBRB. In contrast, the eventual re-organization of TJ complexes at the cell border coincided with restoration of the barrier. A robust inflammatory response was evident a 1 day after IR and progressed to resolution over the 4-week time course. The inflammatory response included a rapid and transient infiltration of granulocytes and Ly6C
+ classical inflammatory monocytes, a slow accumulation of Ly6Cneg monocyte/macrophages, and activation, proliferation, and mobilization of resident microglia. Extravasation of the majority of CD45+ leukocytes occurred from the superficial plexus. The presence of monocyte/macrophages and increased numbers of microglia were sustained until the iBRB was eventually restored. Intervention with minocycline to reverse microglial activation at 1 week after injury promoted early restoration of the iBRB coinciding with decreased expression of mRNAs for the microglial M1 markers TNF-α, IL-1β, and Ptgs2 (Cox-2) and increased expression of secreted serine protease inhibitor Serpina3n mRNA., Conclusions: These results suggest that iBRB restoration occurs as TJ complexes are reorganized and that resolution of inflammation and restoration of the iBRB following retinal IR injury are functionally linked., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
17. Correlation of Retinal Structure and Visual Function Assessments in Mouse Diabetes Models.
- Author
-
Sheskey SR, Antonetti DA, Rentería RC, and Lin CM
- Subjects
- Animals, Diabetes Mellitus, Experimental physiopathology, Diabetic Retinopathy physiopathology, Follow-Up Studies, Male, Mice, Mice, Inbred C57BL, Retina physiopathology, Retinal Vessels physiopathology, Diabetes Mellitus, Experimental diagnosis, Diabetic Retinopathy pathology, Retina pathology, Retinal Vessels pathology, Tomography, Optical Coherence methods, Visual Acuity physiology
- Abstract
Purpose: Diabetic retinopathy results in vision loss with changes to both retinal blood vessels and neural retina. Recent studies have revealed that animal models of diabetes demonstrate early loss of visual function. We explored the time course of retinal change in three different mouse models of diabetes in a longitudinal study using in vivo measures of retinal structure (optical coherence tomography [OCT]) and visual function (optomotor and pupillary responses)., Methods: OCT analysis of retinal microstructure, optokinetic response as a measure of visual acuity, and pupillary response to light stimulation were compared among the db/db, Ins2Akita, and streptozotocin (STZ)-induced mouse models of diabetes at 1.5, 3, 6, and 9 months of diabetes., Results: The db/db, Ins2Akita, and STZ-induced models of diabetes all exhibited vision loss and retinal thinning as disease progressed. Both structural changes and functional measures were significantly correlated with the blood glucose levels. Despite this, vision loss and retinal thinning were not consistently correlated, except for the inner retinal layer thickness at 6 months of diabetes., Conclusions: This longitudinal study compiled structural measures and functional outcome data for type 1 and 2 diabetes mouse models commonly used for diabetes studies and demonstrated an overall decline in retinal-related health in conjunction with weight change and blood glucose alterations. The relationship between the structural change and functional outcome could be correlative but is not necessarily causative, as retinal thinning was not sufficient to explain visual acuity decline.
- Published
- 2021
- Full Text
- View/download PDF
18. Vascular Expression of Permeability-Resistant Occludin Mutant Preserves Visual Function in Diabetes.
- Author
-
Goncalves A, Dreffs A, Lin CM, Sheskey S, Hudson N, Keil J, Campbell M, and Antonetti DA
- Subjects
- Animals, Leukostasis prevention & control, Mice, Mice, Inbred C57BL, Permeability, Phosphorylation, Streptozocin, Vascular Endothelial Growth Factor A physiology, Blood-Retinal Barrier physiology, Diabetes Mellitus, Experimental physiopathology, Diabetic Retinopathy physiopathology, Occludin physiology, Visual Acuity
- Abstract
Diabetic retinopathy is one of the leading causes of vision loss and blindness. Extensive preclinical and clinical evidence exists for both vascular and neuronal pathology. However, the relationship of these changes in the neurovascular unit and impact on vision remains to be determined. Here, we investigate the role of tight junction protein occludin phosphorylation at S490 in modulating barrier properties and its impact on visual function. Conditional vascular expression of the phosphorylation-resistant Ser490 to Ala (S490A) form of occludin preserved tight junction organization and reduced vascular endothelial growth factor (VEGF)-induced permeability and edema formation after intraocular injection. In the retinas of streptozotocin-induced diabetic mice, endothelial-specific expression of the S490A form of occludin completely prevented diabetes-induced permeability to labeled dextran and inhibited leukostasis. Importantly, vascular-specific expression of the occludin mutant completely blocked the diabetes-induced decrease in visual acuity and contrast sensitivity. Together, these results reveal that occludin acts to regulate barrier properties downstream of VEGF in a phosphorylation-dependent manner and that loss of inner blood-retinal barrier integrity induced by diabetes contributes to vision loss., (© 2021 by the American Diabetes Association.)
- Published
- 2021
- Full Text
- View/download PDF
19. Current understanding of the molecular and cellular pathology of diabetic retinopathy.
- Author
-
Antonetti DA, Silva PS, and Stitt AW
- Subjects
- Animals, Diabetic Retinopathy metabolism, Humans, Neurons metabolism, Neurons pathology, Neurovascular Coupling, Signal Transduction, Blood-Retinal Barrier pathology, Diabetic Retinopathy pathology
- Abstract
Diabetes mellitus has profound effects on multiple organ systems; however, the loss of vision caused by diabetic retinopathy might be one of the most impactful in a patient's life. The retina is a highly metabolically active tissue that requires a complex interaction of cells, spanning light sensing photoreceptors to neurons that transfer the electrochemical signal to the brain with support by glia and vascular tissue. Neuronal function depends on a complex inter-dependency of retinal cells that includes the formation of a blood-retinal barrier. This dynamic system is negatively affected by diabetes mellitus, which alters normal cell-cell interactions and leads to profound vascular abnormalities, loss of the blood-retinal barrier and impaired neuronal function. Understanding the normal cell signalling interactions and how they are altered by diabetes mellitus has already led to novel therapies that have improved visual outcomes in many patients. Research highlighted in this Review has led to a new understanding of retinal pathophysiology during diabetes mellitus and has uncovered potential new therapeutic avenues to treat this debilitating disease.
- Published
- 2021
- Full Text
- View/download PDF
20. Updating the Staging System for Diabetic Retinal Disease.
- Author
-
Sun JK, Aiello LP, Abràmoff MD, Antonetti DA, Dutta S, Pragnell M, Levine SR, and Gardner TW
- Subjects
- Diabetic Retinopathy physiopathology, Humans, Retinal Vessels physiopathology, Diabetic Retinopathy diagnosis, Retinal Vessels pathology
- Published
- 2021
- Full Text
- View/download PDF
21. The neuroscience of diabetic retinopathy.
- Author
-
Antonetti DA
- Subjects
- Humans, Neurons, Signal Transduction, Diabetes Mellitus, Diabetic Retinopathy
- Abstract
Diabetic retinopathy remains a leading cause of blindness despite recent advance in therapies. Traditionally, this complication of diabetes was viewed predominantly as a microvascular disease but research has pointed to alterations in ganglion cells, glia, microglia, and photoreceptors as well, often occurring without obvious vascular damage. In neural tissue, the microvasculature and neural tissue form an intimate relationship with the neural tissue providing signaling cues for the vessels to form a distinct barrier that helps to maintain the proper neuronal environment for synaptic signaling. This relationship has been termed the neurovascular unit (NVU). Research is now focused on understanding the cellular and molecular basis of the neurovascular unit and how diabetes alters the normal cellular communications and disrupts the cellular environment contributing to loss of vision in diabetes.
- Published
- 2021
- Full Text
- View/download PDF
22. All-trans-Retinaldehyde Contributes to Retinal Vascular Permeability in Ischemia Reperfusion.
- Author
-
Dreffs A, Lin CM, Liu X, Shanmugam S, Abcouwer SF, Kern TS, and Antonetti DA
- Subjects
- Animals, Cattle, Cell Death, DNA Fragmentation, Dark Adaptation, Electric Impedance, Endothelial Cells drug effects, Enzyme-Linked Immunosorbent Assay, Intraocular Pressure physiology, Mice, Mice, Inbred C57BL, Oxidative Stress drug effects, Phenyl Ethers pharmacology, Propanolamines pharmacology, cis-trans-Isomerases antagonists & inhibitors, Capillary Permeability physiology, Reperfusion Injury metabolism, Retinal Vessels metabolism, Retinaldehyde physiology
- Abstract
Purpose: Extracellular accumulation of all-trans-retinaldehyde (atRAL), a highly reactive visual cycle intermediate, is toxic to cells of the outer retina and contributes to retinal and macular degenerations. However, the contribution of atRAL to retinal capillary function has not been studied. We hypothesized that atRAL released from the outer retina can contribute to retinal vascular permeability. We, therefore, tested the contribution of atRAL to retinal ischemia-reperfusion (IR)-induced vascular permeability., Methods: IR was induced in mice by transient increase in intraocular pressure followed by natural reperfusion. The visual cycle was ablated in the Lrat-/- mice, reduced by dark adaptation or the use of the RPE65 inhibitor and atRAL scavenger emixustat. Accumulation of FITC-BSA was used to assess vascular permeability and DNA fragmentation quantified cell death after IR. Primary bovine retinal endothelial cell (BREC) culture was used to measure the direct effects of atRAL on endothelial permeability and cell death., Results: Inhibition of the visual cycle by Lrat-/-, dark adaptation, or with emixustat, all reduced approximately half of IR induced vascular permeability at 48 hours. An increase in BREC permeability with atRAL coincided with lactate dehydrogenase (LDH) release, a measure of cell death. Both permeability and toxicity were blocked by emixustat., Conclusions: Outer retinal pathology may contribute to vascular permeability by release of atRAL, which can act directly on vascular endothelial cells to alter barrier properties and induce cell death. These studies may have implications for a variety of blinding eye diseases that include outer retinal damage and retinal vascular permeability.
- Published
- 2020
- Full Text
- View/download PDF
23. Synthesis and structure-activity relationships of thieno[2,3-d]pyrimidines as atypical protein kinase C inhibitors to control retinal vascular permeability and cytokine-induced edema.
- Author
-
Liu X, Wilson MW, Liu K, Lee P, Yeomans L, Hagen SE, Lin CM, Wen B, Sun D, White AD, Showalter HD, and Antonetti DA
- Subjects
- Animals, Cytokines metabolism, Dose-Response Relationship, Drug, Edema chemically induced, Edema metabolism, Female, HEK293 Cells, Humans, Mice, Molecular Structure, Protein Kinase C metabolism, Protein Kinase Inhibitors chemical synthesis, Protein Kinase Inhibitors chemistry, Pyrimidines chemical synthesis, Pyrimidines chemistry, Rats, Rats, Long-Evans, Structure-Activity Relationship, Vascular Endothelial Growth Factor A metabolism, Cytokines antagonists & inhibitors, Edema drug therapy, Protein Kinase C antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Pyrimidines pharmacology, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
Studies demonstrate that small molecule targeting of atypical protein kinase C (aPKC) may provide an effective means to control vascular permeability, prevent edema, and reduce inflammation providing novel and important alternatives to anti-VEGF therapies for certain blinding eye diseases. Based on a literature tricyclic thieno[2,3-d]pyrimidine lead (1), an ATP-competitive inhibitor of the aPKC iota (ι) and aPKC zeta (ζ) isoforms, we have synthesized a small series of compounds in 1-2 steps from a readily available chloro intermediate. A single pyridine congener was also made using 2D NMR to assign regiochemistry. Within the parent pyrimidine series, a range of potencies was observed against aPKCζ whereas the pyridine congener was inactive. Selected compounds were also tested for their effect toward VEGF-induced permeability in BREC cells. The most potent of these (7l) was further assayed against the aPKCι isoform and showed a favorable selectivity profile against a panel of 31 kinases, including kinases from the AGC superfamily, with a focus on PKC isoforms and kinases previously shown to affect permeability. Further testing of 7l in a luciferase assay in HEK293 cells showed an ability to prevent TNF-α induced NFκB activation while not having any effect on cell survival. Intravitreal administration of 7l to the eye yielded a complete reduction in permeability in a test to determine whether the compound could block VEGF- and TNFα-induced permeability across the retinal vasculature in a rat model. The compound in mice displayed good microsomal stability and in plasma moderate exposure (AUC and C
max ), low clearance, a long half-life and high oral bioavailability. With IV dosing, higher levels were observed in the brain and eye relative to plasma, with highest levels in the eye by either IV or PO dosing. With a slow oral absorption profile, 7l accumulates in the eye to maintain a high concentration after dosing with higher levels than in plasma. Compound 7l may represent a class of aPKC inhibitors for further investigation., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
24. Norrin restores blood-retinal barrier properties after vascular endothelial growth factor-induced permeability.
- Author
-
Díaz-Coránguez M, Lin CM, Liebner S, and Antonetti DA
- Subjects
- Animals, Blood-Retinal Barrier drug effects, Cattle, Claudin-5 metabolism, Diabetes Mellitus, Experimental chemically induced, Diabetes Mellitus, Experimental pathology, Male, Mitogen-Activated Protein Kinase Kinases metabolism, Rats, Rats, Long-Evans, Retina metabolism, Retinal Vessels cytology, Retinal Vessels metabolism, Signal Transduction drug effects, Tetraspanins genetics, Tetraspanins metabolism, Up-Regulation drug effects, beta Catenin antagonists & inhibitors, beta Catenin metabolism, Blood-Retinal Barrier metabolism, Capillary Permeability drug effects, Eye Proteins pharmacology, Vascular Endothelial Growth Factor A pharmacology
- Abstract
Vascular endothelial growth factor (VEGF) contributes to blood-retinal barrier (BRB) dysfunction in several blinding eye diseases, including diabetic retinopathy. Signaling via the secreted protein norrin through the frizzled class receptor 4 (FZD4)/LDL receptor-related protein 5-6 (LRP5-6)/tetraspanin 12 (TSPAN12) receptor complex is required for developmental vascularization and BRB formation. Here, we tested the hypothesis that norrin restores BRB properties after VEGF-induced vascular permeability in diabetic rats or in animals intravitreally injected with cytokines. Intravitreal co-injection of norrin with VEGF completely ablated VEGF-induced BRB permeability to Evans Blue-albumin. Likewise, 5-month diabetic rats exhibited increased permeability of FITC-albumin, and a single norrin injection restored BRB properties. These results were corroborated in vitro , where co-stimulation of norrin with VEGF or stimulation of norrin after VEGF exposure restored barrier properties, indicated by electrical resistance or 70-kDa RITC-dextran permeability in primary endothelial cell culture. Interestingly, VEGF promoted norrin signaling by increasing the FZD4 co-receptor TSPAN12 at cell membranes in an MAPK/ERK kinase (MEK)/ERK-dependent manner. Norrin signaling through β-catenin was required for BRB restoration, but glycogen synthase kinase 3 α/β (GSK-3α/β) inhibition did not restore BRB properties. Moreover, levels of the tight junction protein claudin-5 were increased with norrin and VEGF or with VEGF alone, but both norrin and VEGF were required for enriched claudin-5 localization at the tight junction. These results suggest that VEGF simultaneously induces vascular permeability and promotes responsiveness to norrin. Norrin, in turn, restores tight junction complex organization and BRB properties in a β-catenin-dependent manner., (© 2020 Díaz-Coránguez et al.)
- Published
- 2020
- Full Text
- View/download PDF
25. Targeting Neurovascular Interaction in Retinal Disorders.
- Author
-
Fu Z, Sun Y, Cakir B, Tomita Y, Huang S, Wang Z, Liu CH, S Cho S, Britton W, S Kern T, Antonetti DA, Hellström A, and E H Smith L
- Subjects
- Animals, Blood Flow Velocity, Humans, Mitochondria metabolism, Reactive Oxygen Species metabolism, Retinal Diseases physiopathology, Retinal Neovascularization physiopathology, Retinal Vessels physiology, Photoreceptor Cells, Vertebrate metabolism, Retina metabolism, Retinal Diseases metabolism, Retinal Neovascularization metabolism, Retinal Vessels metabolism
- Abstract
The tightly structured neural retina has a unique vascular network comprised of three interconnected plexuses in the inner retina (and choroid for outer retina), which provide oxygen and nutrients to neurons to maintain normal function. Clinical and experimental evidence suggests that neuronal metabolic needs control both normal retinal vascular development and pathological aberrant vascular growth. Particularly, photoreceptors, with the highest density of mitochondria in the body, regulate retinal vascular development by modulating angiogenic and inflammatory factors. Photoreceptor metabolic dysfunction, oxidative stress, and inflammation may cause adaptive but ultimately pathological retinal vascular responses, leading to blindness. Here we focus on the factors involved in neurovascular interactions, which are potential therapeutic targets to decrease energy demand and/or to increase energy production for neovascular retinal disorders.
- Published
- 2020
- Full Text
- View/download PDF
26. Organ-On-A-Chip Technologies for Advanced Blood-Retinal Barrier Models.
- Author
-
Ragelle H, Goncalves A, Kustermann S, Antonetti DA, and Jayagopal A
- Subjects
- Animals, Humans, Blood-Retinal Barrier metabolism, Lab-On-A-Chip Devices, Models, Biological, Retina metabolism
- Abstract
The blood-retinal barrier (BRB) protects the retina by maintaining an adequate microenvironment for neuronal function. Alterations of the junctional complex of the BRB and consequent BRB breakdown in disease contribute to a loss of neuronal signaling and vision loss. As new therapeutics are being developed to prevent or restore barrier function, it is critical to implement physiologically relevant in vitro models that recapitulate the important features of barrier biology to improve disease modeling, target validation, and toxicity assessment. New directions in organ-on-a-chip technology are enabling more sophisticated 3-dimensional models with flow, multicellularity, and control over microenvironmental properties. By capturing additional biological complexity, organs-on-chip can help approach actual tissue organization and function and offer additional tools to model and study disease compared with traditional 2-dimensional cell culture. This review describes the current state of barrier biology and barrier function in ocular diseases, describes recent advances in organ-on-a-chip design for modeling the BRB, and discusses the potential of such models for ophthalmic drug discovery and development.
- Published
- 2020
- Full Text
- View/download PDF
27. Tight junction protein occludin regulates progenitor Self-Renewal and survival in developing cortex.
- Author
-
Bendriem RM, Singh S, Aleem AA, Antonetti DA, and Ross ME
- Subjects
- Aneuploidy, Animals, Apoptosis, CRISPR-Cas Systems, Cell Differentiation, Cell Proliferation, Centrosome metabolism, Cerebral Cortex pathology, Disease Models, Animal, Gene Editing, Humans, Mice, Mice, Knockout, Microcephaly genetics, Microcephaly pathology, Microtubules metabolism, Mutagenesis, Mutation, Neurogenesis genetics, Neurogenesis physiology, Occludin genetics, Spindle Apparatus metabolism, Tight Junctions genetics, Cerebral Cortex growth & development, Cerebral Cortex metabolism, Occludin metabolism, Tight Junctions metabolism
- Abstract
Occludin ( OCLN ) mutations cause human microcephaly and cortical malformation. A tight junction component thought absent in neuroepithelium after neural tube closure, OCLN isoform-specific expression extends into corticogenesis. Full-length and truncated isoforms localize to neuroprogenitor centrosomes, but full-length OCLN transiently localizes to plasma membranes while only truncated OCLN continues at centrosomes throughout neurogenesis. Mimicking human mutations, full-length OCLN depletion in mouse and in human CRISPR/Cas9-edited organoids produce early neuronal differentiation, reduced progenitor self-renewal and increased apoptosis. Human neural progenitors were more severely affected, especially outer radial glial cells, which mouse embryonic cortex lacks. Rodent and human mutant progenitors displayed reduced proliferation and prolonged M-phase. OCLN interacted with mitotic spindle regulators, NuMA and RAN, while full-length OCLN loss impaired spindle pole morphology, astral and mitotic microtubule integrity. Thus, early corticogenesis requires full-length OCLN to regulate centrosome organization and dynamics, revealing a novel role for this tight junction protein in early brain development., Competing Interests: RB, SS, AA, DA, MR No competing interests declared, (© 2019, Bendriem et al.)
- Published
- 2019
- Full Text
- View/download PDF
28. Tight Junctions in Cell Proliferation.
- Author
-
Díaz-Coránguez M, Liu X, and Antonetti DA
- Subjects
- Animals, Cell Differentiation physiology, Humans, Signal Transduction physiology, Transcription, Genetic physiology, Cell Proliferation physiology, Tight Junctions physiology
- Abstract
Tight junction (TJ) proteins form a continuous intercellular network creating a barrier with selective regulation of water, ion, and solutes across endothelial, epithelial, and glial tissues. TJ proteins include the claudin family that confers barrier properties, members of the MARVEL family that contribute to barrier regulation, and JAM molecules, which regulate junction organization and diapedesis. In addition, the membrane-associated proteins such as MAGUK family members, i.e., zonula occludens, form the scaffold linking the transmembrane proteins to both cell signaling molecules and the cytoskeleton. Most studies of TJ have focused on the contribution to cell-cell adhesion and tissue barrier properties. However, recent studies reveal that, similar to adherens junction proteins, TJ proteins contribute to the control of cell proliferation. In this review, we will summarize and discuss the specific role of TJ proteins in the control of epithelial and endothelial cell proliferation. In some cases, the TJ proteins act as a reservoir of critical cell cycle modulators, by binding and regulating their nuclear access, while in other cases, junctional proteins are located at cellular organelles, regulating transcription and proliferation. Collectively, these studies reveal that TJ proteins contribute to the control of cell proliferation and differentiation required for forming and maintaining a tissue barrier.
- Published
- 2019
- Full Text
- View/download PDF
29. Pathophysiology of Diabetic Retinopathy: Contribution and Limitations of Laboratory Research.
- Author
-
Kern TS, Antonetti DA, and Smith LEH
- Subjects
- Animals, Diabetic Retinopathy diagnosis, Fundus Oculi, Humans, Diabetic Retinopathy physiopathology, Fluorescein Angiography methods, Research, Retinal Vessels diagnostic imaging, Visual Acuity
- Abstract
Preclinical models of diabetic retinopathy are indispensable in the drug discovery and development of new therapies. They are, however, imperfect facsimiles of diabetic retinopathy in humans. This chapter discusses the advantages, limitations, and physiological and pathological relevance of preclinical models of diabetic retinopathy. The judicious interpretation and extrapolation of data derived from these models to humans and a correspondingly greater emphasis placed on translational medical research in early-stage clinical trials are essential to more successfully inhibit the development and progression of diabetic retinopathy in the future., (© 2019 S. Karger AG, Basel.)
- Published
- 2019
- Full Text
- View/download PDF
30. Inhibition of Atypical Protein Kinase C Reduces Inflammation-Induced Retinal Vascular Permeability.
- Author
-
Lin CM, Titchenell PM, Keil JM, Garcia-Ocaña A, Bolinger MT, Abcouwer SF, and Antonetti DA
- Subjects
- Animals, Capillary Permeability drug effects, Male, Mice, Inbred C57BL, Papilledema chemically induced, Papilledema physiopathology, Rats, Long-Evans, Recombinant Proteins, Reperfusion Injury physiopathology, Retinitis chemically induced, Retinitis physiopathology, Tight Junctions chemistry, Tight Junctions physiology, Tumor Necrosis Factor-alpha pharmacology, Vascular Endothelial Growth Factor A pharmacology, Capillary Permeability physiology, Protein Kinase C antagonists & inhibitors, Retinal Vessels physiology
- Abstract
Changes in permeability of retinal blood vessels contribute to macular edema and the pathophysiology of numerous ocular diseases, including diabetic retinopathy, retinal vein occlusions, and macular degeneration. Vascular endothelial growth factor (VEGF) induces retinal permeability and macular thickening in these diseases. However, inflammatory agents, such as tumor necrosis factor-α (TNF-α), also may drive vascular permeability, specifically in patients unresponsive to anti-VEGF therapy. Recent evidence suggests VEGF and TNF-α induce permeability through distinct mechanisms; however, both require the activation of atypical protein kinase C (aPKC). We provide evidence, using genetic mouse models and therapeutic intervention with small molecules, that inhibition of aPKC prevented or reduced vascular permeability in animal models of retinal inflammation. Expression of a kinase-dead aPKC transgene, driven by a vascular and hematopoietic restricted promoter, reduced retinal vascular permeability in an ischemia-reperfusion model of retinal injury. This effect was recapitulated with a small-molecule inhibitor of aPKC. Expression of the kinase-dead aPKC transgene dramatically reduced the expression of inflammatory factors and blocked the attraction of inflammatory monocytes and granulocytes after ischemic injury. Coinjection of VEGF with TNF-α was sufficient to induce permeability, edema, and retinal inflammation, and treatment with an aPKC inhibitor prevented VEGF/TNF-α-induced permeability. These data suggest that aPKC contributes to inflammation-driven retinal vascular pathology and may be an attractive target for therapeutic intervention., (Copyright © 2018 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
31. Brain endothelial cell junctions after cerebral hemorrhage: Changes, mechanisms and therapeutic targets.
- Author
-
Keep RF, Andjelkovic AV, Xiang J, Stamatovic SM, Antonetti DA, Hua Y, and Xi G
- Subjects
- Animals, Blood-Brain Barrier drug effects, Blood-Brain Barrier metabolism, Brain drug effects, Brain metabolism, Brain pathology, Cerebral Hemorrhage drug therapy, Cerebral Hemorrhage metabolism, Claudin-5 analysis, Claudin-5 metabolism, Endothelial Cells drug effects, Endothelial Cells metabolism, Endothelial Cells pathology, Humans, Intercellular Junctions drug effects, Intercellular Junctions metabolism, Occludin analysis, Occludin metabolism, Zonula Occludens-1 Protein analysis, Zonula Occludens-1 Protein metabolism, Blood-Brain Barrier pathology, Cerebral Hemorrhage pathology, Intercellular Junctions pathology
- Abstract
Vascular disruption is the underlying cause of cerebral hemorrhage, including intracerebral, subarachnoid and intraventricular hemorrhage. The disease etiology also involves cerebral hemorrhage-induced blood-brain barrier (BBB) disruption, which contributes an important component to brain injury after the initial cerebral hemorrhage. BBB loss drives vasogenic edema, allows leukocyte extravasation and may lead to the entry of potentially neurotoxic and vasoactive compounds into brain. This review summarizes current information on changes in brain endothelial junction proteins in response to cerebral hemorrhage (and clot-related factors), the mechanisms underlying junction modification and potential therapeutic targets to limit BBB disruption and, potentially, hemorrhage occurrence. It also addresses advances in the tools that are now available for assessing changes in junctions after cerebral hemorrhage and the potential importance of such junction changes. Recent studies suggest post-translational modification, conformational change and intracellular trafficking of junctional proteins may alter barrier properties. Understanding how cerebral hemorrhage alters BBB properties beyond changes in tight junction protein loss may provide important therapeutic insights to prevent BBB dysfunction and restore normal function.
- Published
- 2018
- Full Text
- View/download PDF
32. Retinal pH and Acid Regulation During Metabolic Acidosis.
- Author
-
Dreffs A, Henderson D, Dmitriev AV, Antonetti DA, and Linsenmeier RA
- Subjects
- Acidosis genetics, Acidosis physiopathology, Animals, Blotting, Western, Disease Models, Animal, Electroretinography, Eye Proteins biosynthesis, Eye Proteins genetics, Gene Expression Regulation, Hydrogen-Ion Concentration, Immunohistochemistry, Male, RNA genetics, Rats, Rats, Long-Evans, Retina physiopathology, Reverse Transcriptase Polymerase Chain Reaction, Sodium-Hydrogen Exchangers biosynthesis, Sodium-Hydrogen Exchangers genetics, Acidosis metabolism, Retina metabolism
- Abstract
Purpose: Changes in retinal pH may contribute to a variety of eye diseases. To study the effect of acidosis alone, we induced systemic metabolic acidosis and hypothesized that the retina would respond with altered expression of genes involved in acid/base regulation., Methods: Systemic metabolic acidosis was induced in Long-Evans rats for up to 2 weeks by adding NH
4 Cl to the drinking water. After 2 weeks, venous pH was 7.25 ± 0.08 (SD) and [HCO3 - ] was 21.4 ± 4.6 mM in acidotic animals; pH was 7.41 ± 0.03 and [HCO3 - ] was 30.5 ± 1.0 mM in controls. Retinal mRNAs were quantified by quantitative reverse transcription polymerase chain reaction. Protein was quantified with Western blots and localized by confocal microscopy. Retinal [H+ ]o was measured in vivo with pH microelectrodes in animals subjected to metabolic acidosis and in controls., Results: NH4 Cl in drinking water or given intravenous was effective in acidifying the retina. Cariporide, a blocker of Na+ /H+ exchange, further acidified the retina. Metabolic acidosis for 2 weeks led to increases of 40-100% in mRNA for carbonic anhydrase isoforms II (CA-II) and XIV (CA-XIV) and acid-sensing ion channels 1 and 4 (ASIC1 and ASIC4) (all p < 0.005). Expression of anion exchange protein 3 (AEP-3) and Na+ /H+ exchanger (NHE)-1 also increased by ≥50% (both p < 0.0001). Changes were similar after 1 week of acidosis. Protein for AEP-3 doubled. NHE-1 co-localized with vascular markers, particularly in the outer plexiform layer. CA-II was located in the neural parenchyma of the ganglion cell layer and diffusely in the rest of the inner retina., Conclusions: The retina responds to systemic acidosis with increased expression of proton and bicarbonate exchangers, carbonic anhydrase, and ASICs. While responses to acidosis are usually associated with renal regulation, these studies suggest that the retina responds to changes in local pH presumably to control its acid/base environment in response to systemic acidosis.- Published
- 2018
- Full Text
- View/download PDF
33. ELOVL4-Mediated Production of Very Long-Chain Ceramides Stabilizes Tight Junctions and Prevents Diabetes-Induced Retinal Vascular Permeability.
- Author
-
Kady NM, Liu X, Lydic TA, Syed MH, Navitskaya S, Wang Q, Hammer SS, O'Reilly S, Huang C, Seregin SS, Amalfitano A, Chiodo VA, Boye SL, Hauswirth WW, Antonetti DA, and Busik JV
- Subjects
- Animals, Cattle, Claudin-5 metabolism, Diabetes Mellitus, Experimental complications, Diabetes Mellitus, Experimental metabolism, Diabetic Retinopathy etiology, Diabetic Retinopathy genetics, Diabetic Retinopathy metabolism, Endothelial Cells ultrastructure, Humans, Interleukin-1beta metabolism, Mice, Occludin metabolism, Retina metabolism, Retinal Vessels ultrastructure, Tight Junctions ultrastructure, Vascular Endothelial Growth Factor A metabolism, Zonula Occludens-1 Protein metabolism, Blood-Retinal Barrier metabolism, Capillary Permeability genetics, Ceramides metabolism, Endothelial Cells metabolism, Eye Proteins genetics, Eye Proteins metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Retinal Vessels metabolism, Tight Junctions metabolism
- Abstract
Tight junctions (TJs) involve close apposition of transmembrane proteins between cells. Although TJ proteins have been studied in detail, the role of lipids is largely unknown. We addressed the role of very long-chain (VLC ≥26) ceramides in TJs using diabetes-induced loss of the blood-retinal barrier as a model. VLC fatty acids that incorporate into VLC ceramides are produced by elongase elongation of very long-chain fatty acids protein 4 (ELOVL4). ELOVL4 is significantly reduced in the diabetic retina. Overexpression of ELOVL4 significantly decreased basal permeability, inhibited vascular endothelial growth factor (VEGF)- and interleukin-1β-induced permeability, and prevented VEGF-induced decrease in occludin expression and border staining of TJ proteins ZO-1 and claudin-5. Intravitreal delivery of AAV2-hELOVL4 reduced diabetes-induced increase in vascular permeability. Ultrastructure and lipidomic analysis revealed that ω-linked acyl-VLC ceramides colocalize with TJ complexes. Overall, normalization of retinal ELOVL4 expression could prevent blood-retinal barrier dysregulation in diabetic retinopathy through an increase in VLC ceramides and stabilization of TJs., (© 2018 by the American Diabetes Association.)
- Published
- 2018
- Full Text
- View/download PDF
34. The EPAC-Rap1 pathway prevents and reverses cytokine-induced retinal vascular permeability.
- Author
-
Ramos CJ, Lin C, Liu X, and Antonetti DA
- Subjects
- Acetylcysteine metabolism, Animals, Erythromycin metabolism, Humans, Receptors, Vascular Endothelial Growth Factor metabolism, Signal Transduction drug effects, Acetylcysteine analogs & derivatives, Capillary Permeability drug effects, Cytokines pharmacology, Erythromycin analogs & derivatives, Retina drug effects, Retina metabolism, rap1 GTP-Binding Proteins metabolism
- Abstract
Increased retinal vascular permeability contributes to macular edema, a leading cause of vision loss in eye pathologies such as diabetic retinopathy, age-related macular degeneration, and central retinal vein occlusions. Pathological changes in vascular permeability are driven by growth factors such as VEGF and pro-inflammatory cytokines such as TNF-α. Identifying the pro-barrier mechanisms that block vascular permeability and restore the blood-retinal barrier (BRB) may lead to new therapies. The cAMP-dependent guanine nucleotide exchange factor (EPAC) exchange-protein directly activated by cAMP promotes exchange of GTP in the small GTPase Rap1. Rap1 enhances barrier properties in human umbilical endothelial cells by promoting adherens junction assembly. We hypothesized that the EPAC-Rap1 signaling pathway may regulate the tight junction complex of the BRB and may restore barrier properties after cytokine-induced permeability. Here, we show that stimulating EPAC or Rap1 activation can prevent or reverse VEGF- or TNF-α-induced permeability in cell culture and in vivo Moreover, EPAC activation inhibited VEGF receptor (VEGFR) signaling through the Ras/MEK/ERK pathway. We also found that Rap1B knockdown or an EPAC antagonist increases endothelial permeability and that VEGF has no additive effect, suggesting a common pathway. Furthermore, GTP-bound Rap1 promoted tight junction assembly, and loss of Rap1B led to loss of junctional border organization. Collectively, our results indicate that the EPAC-Rap1 pathway helps maintain basal barrier properties in the retinal vascular endothelium and activation of the EPAC-Rap1 pathway may therefore represent a potential therapeutic strategy to restore the BRB., (© 2018 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
35. Cell autonomous sonic hedgehog signaling contributes to maintenance of retinal endothelial tight junctions.
- Author
-
Díaz-Coránguez M, Chao DL, Salero EL, Goldberg JL, and Antonetti DA
- Subjects
- Animals, Blotting, Western, Cattle, Cell Membrane Permeability drug effects, Cells, Cultured, Disease Models, Animal, Electric Impedance, Endothelial Cells drug effects, Retina drug effects, Signal Transduction drug effects, Tight Junctions drug effects, Vascular Endothelial Growth Factor A pharmacology, Veratrum Alkaloids pharmacology, Endothelial Cells physiology, Hedgehog Proteins physiology, Retina physiology, Signal Transduction physiology, Tight Junctions physiology
- Published
- 2017
- Full Text
- View/download PDF
36. The inner blood-retinal barrier: Cellular basis and development.
- Author
-
Díaz-Coránguez M, Ramos C, and Antonetti DA
- Subjects
- Animals, Capillary Permeability physiology, Humans, Retinal Vessels physiology, Blood-Retinal Barrier physiology, Diabetic Retinopathy physiopathology
- Abstract
The blood-retinal barrier (BRB) regulates transport across retinal capillaries maintaining proper neural homeostasis and protecting the neural tissue from potential blood borne toxicity. Loss of the BRB contributes to the pathophysiology of a number of blinding retinal diseases including diabetic retinopathy. In this review, we address the basis of the BRB, including the molecular mechanisms that regulate flux across the retinal vascular bed. The routes of transcellular and paracellular flux are described as well as alterations in these pathways in response to permeabilizing agents in diabetes. Finally, we provide information on exciting new studies that help to elucidate the process of BRB development or barriergenesis and how understanding this process may lead to new opportunities for barrier restoration in diabetic retinopathy., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
37. The role of small GTPases and EPAC-Rap signaling in the regulation of the blood-brain and blood-retinal barriers.
- Author
-
Ramos CJ and Antonetti DA
- Subjects
- Acetylcysteine analogs & derivatives, Acetylcysteine metabolism, Animals, Erythromycin analogs & derivatives, Erythromycin metabolism, Humans, Monomeric GTP-Binding Proteins metabolism, Blood-Brain Barrier metabolism, Blood-Retinal Barrier metabolism, Signal Transduction
- Abstract
Maintenance and regulation of the vascular endothelial cell junctional complex is critical for proper barrier function of the blood-brain barrier (BBB) and the highly related blood-retinal barrier (BRB) that help maintain proper neuronal environment. Recent research has demonstrated that the junctional complex is actively maintained and can be dynamically regulated. Studies focusing on the mechanisms of barrier formation, maintenance, and barrier disruption have been of interest to understanding development of the BBB and BRB and identifying a means for therapeutic intervention for diseases ranging from brain tumors and dementia to blinding eye diseases. Research has increasingly revealed that small GTPases play a critical role in both barrier formation and disruption mechanisms. This review will summarize the current data on small GTPases in barrier regulation with an emphasis on the EPAC-Rap1 signaling pathway to Rho in endothelial barriers, as well as explore its potential involvement in paracellular flux and transcytosis regulation.
- Published
- 2017
- Full Text
- View/download PDF
38. Moving Past Anti-VEGF: Novel Therapies for Treating Diabetic Retinopathy.
- Author
-
Bolinger MT and Antonetti DA
- Subjects
- Animals, Bevacizumab therapeutic use, Diabetic Retinopathy metabolism, Drug Therapy methods, Drug Therapy trends, Humans, Kallikreins metabolism, Tumor Necrosis Factor-alpha metabolism, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor A metabolism, Adalimumab therapeutic use, Diabetic Retinopathy drug therapy, Kallikreins antagonists & inhibitors, Peptides therapeutic use, Tumor Necrosis Factor-alpha antagonists & inhibitors
- Abstract
Diabetic retinopathy is the leading cause of blindness in working age adults, and is projected to be a significant future health concern due to the rising incidence of diabetes. The recent advent of anti-vascular endothelial growth factor (VEGF) antibodies has revolutionized the treatment of diabetic retinopathy but a significant subset of patients fail to respond to treatment. Accumulating evidence indicates that inflammatory cytokines and chemokines other than VEGF may contribute to the disease process. The current review examines the presence of non-VEGF cytokines in the eyes of patients with diabetic retinopathy and highlights mechanistic pathways in relevant animal models. Finally, novel drug targets including components of the kinin-kallikrein system and emerging treatments such as anti-HPTP (human protein tyrosine phosphatase) β antibodies are discussed. Recognition of non-VEGF contributions to disease pathogenesis may lead to novel therapeutics to enhance existing treatments for patients who do not respond to anti-VEGF therapies., Competing Interests: David A. Antonetti has grant support from NovoNordisk and Unity Bioscience Mark T. Bolinger declares no conflict of interest. These funding sources had no role in the development or writing of this review.
- Published
- 2016
- Full Text
- View/download PDF
39. Occludin S490 Phosphorylation Regulates Vascular Endothelial Growth Factor-Induced Retinal Neovascularization.
- Author
-
Liu X, Dreffs A, Díaz-Coránguez M, Runkle EA, Gardner TW, Chiodo VA, Hauswirth WW, and Antonetti DA
- Subjects
- Animals, Blood-Retinal Barrier metabolism, Blotting, Western, Cattle, Humans, Immunohistochemistry, Mice, Mice, Transgenic, Phosphorylation, Occludin metabolism, Retinal Neovascularization metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
Occludin is a transmembrane tight junction protein that contributes to diverse cellular functions, including control of barrier properties, cell migration, and proliferation. Vascular endothelial growth factor (VEGF) induces phosphorylation of occludin at S490, which is required for VEGF-induced endothelial permeability. Herein, we demonstrate that occludin S490 phosphorylation also regulates VEGF-induced retinal endothelial cell proliferation and neovascularization. Using a specific antibody, phospho-occludin was located in centrosomes in endothelial cell cultures, animal models, and human surgical samples of retinal neovessels. Occludin S490 phosphorylation was found to increase with endothelial tube formation in vitro and in vivo during retinal neovascularization after induction of VEGF expression. More important, expression of occludin mutated at S490 to Ala, completely inhibited angiogenesis in cell culture models and in vivo. Collectively, these data suggest a novel role for occludin in regulation of endothelial proliferation and angiogenesis in a phosphorylation-dependent manner. These findings may lead to methods of regulating pathological neovascularization by specifically targeting endothelial cell proliferation., (Copyright © 2016 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
40. Occludin S471 Phosphorylation Contributes to Epithelial Monolayer Maturation.
- Author
-
Bolinger MT, Ramshekar A, Waldschmidt HV, Larsen SD, Bewley MC, Flanagan JM, and Antonetti DA
- Subjects
- Animals, Cell Proliferation, Dogs, Epithelial Cells metabolism, Humans, Madin Darby Canine Kidney Cells, Occludin chemistry, Occludin genetics, Phosphorylation, Protein Domains, Epithelial Cells cytology, G-Protein-Coupled Receptor Kinases metabolism, Occludin metabolism, Serine metabolism, Tight Junctions metabolism
- Abstract
Multiple organ systems require epithelial barriers for normal function, and barrier loss is a hallmark of diseases ranging from inflammation to epithelial cancers. However, the molecular processes regulating epithelial barrier maturation are not fully elucidated. After contact, epithelial cells undergo size-reductive proliferation and differentiate, creating a dense, highly ordered monolayer with high resistance barriers. We provide evidence that the tight junction protein occludin contributes to the regulation of epithelial cell maturation upon phosphorylation of S471 in its coiled-coil domain. Overexpression of a phosphoinhibitory occludin S471A mutant prevents size-reductive proliferation and subsequent tight junction maturation in a dominant manner. Inhibition of cell proliferation in cell-contacted but immature monolayers recapitulated this phenotype. A kinase screen identified G-protein-coupled receptor kinases (GRKs) targeting S471, and GRK inhibitors delayed epithelial packing and junction maturation. We conclude that occludin contributes to the regulation of size-reductive proliferation and epithelial cell maturation in a phosphorylation-dependent manner., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
41. Protective Effect of a GLP-1 Analog on Ischemia-Reperfusion Induced Blood-Retinal Barrier Breakdown and Inflammation.
- Author
-
Gonçalves A, Lin CM, Muthusamy A, Fontes-Ribeiro C, Ambrósio AF, Abcouwer SF, Fernandes R, and Antonetti DA
- Subjects
- Animals, Cattle, Cells, Cultured, Disease Models, Animal, Exenatide, Immunoblotting, Immunohistochemistry, Incretins pharmacology, Inflammation metabolism, Ischemia etiology, Ischemia metabolism, Male, Rats, Rats, Long-Evans, Reperfusion Injury metabolism, Retinal Diseases etiology, Retinal Diseases metabolism, Blood-Retinal Barrier drug effects, Glucagon-Like Peptide 1 analogs & derivatives, Inflammation prevention & control, Ischemia prevention & control, Peptides pharmacology, Reperfusion Injury complications, Retinal Diseases prevention & control, Venoms pharmacology
- Abstract
Purpose: Inflammation associated with blood-retinal barrier (BRB) breakdown is a common feature of several retinal diseases. Therefore, the development of novel nonsteroidal anti-inflammatory approaches may provide important therapeutic options. Previous studies demonstrated that inhibition of dipeptidyl peptidase-IV, the enzyme responsible for the degradation of glucagon-like peptide-1 (GLP-1), led to insulin-independent prevention of diabetes-induced increases in BRB permeability, suggesting that incretin-based drugs may have beneficial pleiotropic effects in the retina. In the current study, the barrier protective and anti-inflammatory properties of exendin-4 (Ex-4), an analog of GLP-1, after ischemia-reperfusion (IR) injury were examined., Methods: Ischemia-reperfusion injury was induced in rat retinas by increasing the intraocular pressure for 45 minutes followed by 48 hours of reperfusion. Rats were treated with Ex-4 prior to and following IR. Blood-retinal barrier permeability was assessed by Evans blue dye leakage. Retinal inflammatory gene expression and leukocytic infiltration were measured by qRT-PCR and immunofluorescence, respectively. A microglial cell line was used to determine the effects of Ex-4 on lipopolysaccharide (LPS)-induced inflammatory response., Results: Exendin-4 dramatically reduced the BRB permeability induced by IR injury, which was associated with suppression of inflammatory gene expression. Moreover, in vitro studies showed that Ex-4 also reduced the inflammatory response to LPS and inhibited NF-κB activation., Conclusions: The present work suggests that Ex-4 can prevent IR injury-induced BRB breakdown and inflammation through inhibition of inflammatory cytokine production by activated microglia and may provide a novel option for therapeutic intervention in diseases involving retinal inflammation.
- Published
- 2016
- Full Text
- View/download PDF
42. Algorithm for the Measure of Vitreous Hyperreflective Foci in Optical Coherence Tomographic Scans of Patients With Diabetic Macular Edema.
- Author
-
Korot E, Comer G, Steffens T, and Antonetti DA
- Subjects
- Adult, Aged, Female, Glycated Hemoglobin metabolism, Humans, Imaging, Three-Dimensional, Male, Middle Aged, Reproducibility of Results, Retrospective Studies, Algorithms, Diabetic Retinopathy diagnosis, Macular Edema diagnosis, Tomography, Optical Coherence methods, Vitreous Body pathology
- Abstract
Importance: Developing a noninvasive measure of diabetic retinopathy disease progression may provide physicians with information needed for patient-specific intervention., Objective: To develop an algorithm to measure vitreous hyperreflective foci (VHRF) from standard, 3-dimensional optical coherence tomographic (OCT) images in an unbiased manner., Design, Setting, and Participants: We retrospectively analyzed OCT scans from 97 patients who were evaluated at the Kellogg Eye Center, University of Michigan. Patients with diabetes mellitus without signs of retinopathy (n = 9) and patients with diabetic macular edema (DME) (n = 31) were compared with healthy control participants (n = 37). The algorithm was used to determine whether the VHRF score is associated with DME and may serve as a noninvasive measure of inflammation. The study was conducted from November 14, 2011, to August 5, 2015. Data analysis was performed from May 15, 2014, to August 13, 2015., Main Outcomes and Measures: An algorithm was developed to enhance the vitreous imaging from OCT to allow automated quantification of VHRF and calculation of a VHRF score. This score was compared between the healthy control, diabetes without retinopathy, and DME groups., Results: In the 97 scans evaluated, VHRF scores, reported as mean (SD), were increased in patients with DME by 2.95-fold (5.60 [8.65]) compared with healthy controls (1.90 [3.42]; 95% CI, 0.75-7.45; P = .012) and by 6.83-fold compared with patients with diabetes without retinopathy (0.82 [1.26]; 95% CI, 1.46-8.82; P = .005)., Conclusions and Relevance: Scores obtained using the VHRF algorithm may be obtained from OCT images that include the vitreous and could provide a rapid, noninvasive clinical correlate for ocular inflammation. Higher VHRF scores in patients with DME compared with controls and diabetic patients without retinopathy warrant further population-based and longitudinal studies to help determine the value of the VHRF score in selecting therapeutic intervention.
- Published
- 2016
- Full Text
- View/download PDF
43. Retinylamine Benefits Early Diabetic Retinopathy in Mice.
- Author
-
Liu H, Tang J, Du Y, Lee CA, Golczak M, Muthusamy A, Antonetti DA, Veenstra AA, Amengual J, von Lintig J, Palczewski K, and Kern TS
- Subjects
- Acyltransferases deficiency, Acyltransferases metabolism, Animals, Cell Separation, Diabetic Retinopathy blood, Diabetic Retinopathy pathology, Diabetic Retinopathy physiopathology, Diterpenes administration & dosage, Diterpenes chemistry, Diterpenes pharmacology, Dose-Response Relationship, Drug, Endothelial Cells metabolism, Glucose metabolism, Inflammation pathology, Leukocytes metabolism, Male, Mice, Inbred C57BL, Oxidative Stress drug effects, Permeability, Photoreceptor Cells, Vertebrate metabolism, Retina drug effects, Retina pathology, Retina physiopathology, Superoxides metabolism, Diabetic Retinopathy drug therapy, Diterpenes therapeutic use
- Abstract
Recent evidence suggests an important role for outer retinal cells in the pathogenesis of diabetic retinopathy (DR). Here we investigated the effect of the visual cycle inhibitor retinylamine (Ret-NH2) on the development of early DR lesions. Wild-type (WT) C57BL/6J mice (male, 2 months old when diabetes was induced) were made diabetic with streptozotocin, and some were given Ret-NH2 once per week. Lecithin-retinol acyltransferase (LRAT)-deficient mice and P23H mutant mice were similarly studied. Mice were euthanized after 2 (WT and Lrat(-/-)) and 8 months (WT) of study to assess vascular histopathology, accumulation of albumin, visual function, and biochemical and physiological abnormalities in the retina. Non-retinal effects of Ret-NH2 were examined in leukocytes treated in vivo. Superoxide generation and expression of inflammatory proteins were significantly increased in retinas of mice diabetic for 2 or 8 months, and the number of degenerate retinal capillaries and accumulation of albumin in neural retina were significantly increased in mice diabetic for 8 months compared with nondiabetic controls. Administration of Ret-NH2 once per week inhibited capillary degeneration and accumulation of albumin in the neural retina, significantly reducing diabetes-induced retinal superoxide and expression of inflammatory proteins. Superoxide generation also was suppressed in Lrat(-/-) diabetic mice. Leukocytes isolated from diabetic mice treated with Ret-NH2 caused significantly less cytotoxicity to retinal endothelial cells ex vivo than did leukocytes from control diabetics. Administration of Ret-NH2 once per week significantly inhibited the pathogenesis of lesions characteristic of early DR in diabetic mice. The visual cycle constitutes a novel target for inhibition of DR., (© 2015 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2015
- Full Text
- View/download PDF
44. Adrenergic and serotonin receptors affect retinal superoxide generation in diabetic mice: relationship to capillary degeneration and permeability.
- Author
-
Du Y, Cramer M, Lee CA, Tang J, Muthusamy A, Antonetti DA, Jin H, Palczewski K, and Kern TS
- Subjects
- Animals, Capillaries metabolism, Cells, Cultured, Cyclic AMP metabolism, Diabetes Mellitus, Experimental physiopathology, Diabetic Retinopathy etiology, Diabetic Retinopathy metabolism, Male, Mice, Mice, Inbred C57BL, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Receptors, Adrenergic genetics, Receptors, Serotonin genetics, Retina cytology, Retina metabolism, Retinal Degeneration etiology, Retinal Degeneration metabolism, Reverse Transcriptase Polymerase Chain Reaction, Capillaries pathology, Diabetes Mellitus, Experimental complications, Diabetic Retinopathy pathology, Receptors, Adrenergic metabolism, Receptors, Serotonin metabolism, Retinal Degeneration pathology, Superoxides metabolism
- Abstract
Reactive oxygen species play an important role in the pathogenesis of diabetic retinopathy. We studied the role of adrenergic and serotonin receptors in the generation of superoxide by retina and 661W retinal cells in high glucose and of the α1-adrenergic receptor (AR) on vascular lesions of the retinopathy in experimentally diabetic C57Bl/6J mice (and controls) after 2 and 8 months. Compared with 5 mM glucose, incubating cells or retinal explants in 30 mM glucose induced superoxide generation. This response was reduced or ablated by pharmacologic inhibition of the α1-AR (a Gq-coupled receptor) or Gs-coupled serotonin (5-HT2, 5-HT4, 5-HT6, and 5-HT7) receptors or by activation of the Gi-coupled α2-AR. In elevated glucose, the α1-AR produced superoxide via phospholipase C, inositol triphosphate-induced Ca(2+) release, and NADPH oxidase, and pharmacologic inhibition of these reactions prevented the superoxide increase. Generation of retinal superoxide, expression of proinflammatory proteins, and degeneration of retinal capillaries in diabetes all were significantly inhibited with daily doxazosin or apocynin (inhibitors of α1-AR and NADPH oxidase, respectively), but increased vascular permeability was not significantly affected. Adrenergic receptors, and perhaps other GPCRs, represent novel targets for inhibiting the development of important features of diabetic retinopathy., (© FASEB.)
- Published
- 2015
- Full Text
- View/download PDF
45. Ocular Anti-VEGF therapy for diabetic retinopathy: the role of VEGF in the pathogenesis of diabetic retinopathy.
- Author
-
Simó R, Sundstrom JM, and Antonetti DA
- Subjects
- Angiogenesis Inhibitors administration & dosage, Angiogenesis Inhibitors adverse effects, Humans, Angiogenesis Inhibitors therapeutic use, Diabetic Retinopathy drug therapy, Diabetic Retinopathy metabolism, Intravitreal Injections methods, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor A physiology
- Abstract
Diabetic retinopathy is the leading cause of visual impairment and preventable blindness, and represents a significant socioeconomic cost for health care systems worldwide. Therefore, new approaches beyond current standards of diabetes care are needed. Based on the crucial pathogenic role of vascular endothelial growth factor (VEGF) in the development of diabetic macular edema (DME), intravitreal anti-VEGF agents have emerged as new treatments. To provide an understanding of the rationale for use and clinical efficacy of anti-VEGF treatment, we examine this topic in a two-part Bench to Clinic narrative. In the Bench narrative, we provide an overview of the role of VEGF in the pathogenesis of diabetic retinopathy, the molecular characteristics of anti-VEGF agents currently used, and future perspectives and challenges in this area. In the Clinic narrative that follows our contribution, Cheung et al. provide an overview of the current evidence from clinical trials on anti-VEGF therapy for diabetic retinopathy.
- Published
- 2014
- Full Text
- View/download PDF
46. Ischemia-reperfusion injury induces occludin phosphorylation/ubiquitination and retinal vascular permeability in a VEGFR-2-dependent manner.
- Author
-
Muthusamy A, Lin CM, Shanmugam S, Lindner HM, Abcouwer SF, and Antonetti DA
- Subjects
- Animals, Blood-Retinal Barrier metabolism, Blood-Retinal Barrier pathology, Blotting, Western, Ischemia pathology, Ischemia physiopathology, Male, Phosphorylation, Rats, Rats, Long-Evans, Reperfusion Injury pathology, Reperfusion Injury physiopathology, Ubiquitination, Capillary Permeability physiology, Ischemia metabolism, Occludin metabolism, Reperfusion Injury metabolism, Retina pathology, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Retinal ischemia-reperfusion (IR) induces neurodegenaration as well as blood-retinal barrier (BRB) breakdown causing vascular permeability. Whereas the neuronal death has been extensively studied, the molecular mechanisms related to BRB breakdown in IR injury remain poorly understood. In this study, we investigated the early changes in tight junctional (TJ) proteins in response to IR injury. Ischemia-reperfusion injury was induced in male rat retinas by increasing the intraocular pressure for 45 minutes followed by natural reperfusion. The results demonstrate that IR injury induced occludin Ser490 phosphorylation and ubiquitination within 15 minutes of reperfusion with subsequent vascular permeability. Immunohistochemical analysis revealed a rapid increase in occludin Ser490 phosphorylation and loss of Zonula occludens-1 (ZO-1) protein, particularly in arterioles. Ischemia-reperfusion injury also rapidly induced the activation and phosphorylation of vascular endothelial growth factor receptor-2 (VEGFR-2) at tyrosine 1175. Blocking vascular endothelial growth factor (VEGF) function by intravitreal injection of bevacizumab prevented VEGFR-2 activation, occludin phosphorylation, and vascular permeability. These studies suggest a novel mechanism of occludin Ser490 phosphorylation and ubiquitination downstream of VEGFR2 activation associated with early IR-induced vascular permeability.
- Published
- 2014
- Full Text
- View/download PDF
47. Mechanism of metabolic stroke and spontaneous cerebral hemorrhage in glutaric aciduria type I.
- Author
-
Zinnanti WJ, Lazovic J, Housman C, Antonetti DA, Koeller DM, Connor JR, and Steinman L
- Subjects
- Amino Acid Metabolism, Inborn Errors genetics, Amino Acid Metabolism, Inborn Errors pathology, Animals, Brain metabolism, Brain pathology, Brain ultrastructure, Brain Diseases, Metabolic genetics, Brain Diseases, Metabolic pathology, Capillaries pathology, Disease Models, Animal, Glial Fibrillary Acidic Protein metabolism, Glutaryl-CoA Dehydrogenase genetics, Magnetic Resonance Angiography, Mice, Mice, Inbred C57BL, Mice, Transgenic, Microscopy, Electron, Occludin metabolism, Statistics, Nonparametric, Amino Acid Metabolism, Inborn Errors complications, Brain Diseases, Metabolic complications, Cerebral Hemorrhage etiology, Glutaryl-CoA Dehydrogenase deficiency, Stroke etiology
- Abstract
Background: Metabolic stroke is the rapid onset of lasting central neurological deficit associated with decompensation of an underlying metabolic disorder. Glutaric aciduria type I (GA1) is an inherited disorder of lysine and tryptophan metabolism presenting with metabolic stroke in infancy. The clinical presentation includes bilateral striatal necrosis and spontaneous subdural and retinal hemorrhages, which has been frequently misdiagnosed as non-accidental head trauma. The mechanisms underlying metabolic stroke and spontaneous hemorrhage are poorly understood., Results: Using a mouse model of GA1, we show that metabolic stroke progresses in the opposite sequence of ischemic stroke, with initial neuronal swelling and vacuole formation leading to cerebral capillary occlusion. Focal regions of cortical followed by striatal capillaries are occluded with shunting to larger non-exchange vessels leading to early filling and dilation of deep cerebral veins. Blood-brain barrier breakdown was associated with displacement of tight-junction protein Occludin., Conclusion: Together the current findings illuminate the pathophysiology of metabolic stroke and vascular compromise in GA1, which may translate to other neurometabolic disorders presenting with stroke.
- Published
- 2014
- Full Text
- View/download PDF
48. Minocycline prevents retinal inflammation and vascular permeability following ischemia-reperfusion injury.
- Author
-
Abcouwer SF, Lin CM, Shanmugam S, Muthusamy A, Barber AJ, and Antonetti DA
- Subjects
- Animals, Blood-Retinal Barrier pathology, Capillary Permeability drug effects, Fluorescent Antibody Technique, Gene Expression drug effects, Male, Nerve Degeneration pathology, Rats, Rats, Sprague-Dawley, Real-Time Polymerase Chain Reaction, Retina drug effects, Blood-Retinal Barrier drug effects, Inflammation pathology, Minocycline pharmacology, Neuroprotective Agents pharmacology, Reperfusion Injury pathology, Retina pathology
- Abstract
Background: Many retinal diseases are associated with vascular dysfunction accompanied by neuroinflammation. We examined the ability of minocycline (Mino), a tetracycline derivative with anti-inflammatory and neuroprotective properties, to prevent vascular permeability and inflammation following retinal ischemia-reperfusion (IR) injury, a model of retinal neurodegeneration with breakdown of the blood-retinal barrier (BRB)., Methods: Male Sprague-Dawley rats were subjected to 45 min of pressure-induced retinal ischemia, with the contralateral eye serving as control. Rats were treated with Mino prior to and following IR. At 48 h after reperfusion, retinal gene expression, cellular inflammation, Evan's blue dye leakage, tight junction protein organization, caspase-3 activation, and DNA fragmentation were measured. Cellular inflammation was quantified by flow-cytometric evaluation of retinal tissue using the myeloid marker CD11b and leukocyte common antigen CD45 to differentiate and quantify CD11b+/CD45low microglia, CD11b+/CD45hi myeloid leukocytes and CD11bneg/CD45hi lymphocytes. Major histocompatibility complex class II (MHCII) immunoreactivity was used to determine the inflammatory state of these cells., Results: Mino treatment significantly inhibited IR-induced retinal vascular permeability and disruption of tight junction organization. Retinal IR injury significantly altered mRNA expression for 21 of 25 inflammation- and gliosis-related genes examined. Of these, Mino treatment effectively attenuated IR-induced expression of lipocalin 2 (LCN2), serpin peptidase inhibitor clade A member 3 N (SERPINA3N), TNF receptor superfamily member 12A (TNFRSF12A), monocyte chemoattractant-1 (MCP-1, CCL2) and intercellular adhesion molecule-1 (ICAM-1). A marked increase in leukostasis of both myeloid leukocytes and lymphocytes was observed following IR. Mino treatment significantly reduced retinal leukocyte numbers following IR and was particularly effective in decreasing the appearance of MHCII+ inflammatory leukocytes. Surprisingly, Mino did not significantly inhibit retinal cell death in this model., Conclusions: IR induces a retinal neuroinflammation within hours of reperfusion characterized by inflammatory gene expression, leukocyte adhesion and invasion, and vascular permeability. Despite Mino significantly inhibiting these responses, it failed to block neurodegeneration.
- Published
- 2013
- Full Text
- View/download PDF
49. Polychromatic angiography for the assessment of VEGF-induced BRB dysfunction in the rabbit retina.
- Author
-
Tari SR, Youssif M, Samson CM, Harris RL, Lin CM, Kompella UB, Antonetti DA, and Barile GR
- Subjects
- Animals, Disease Models, Animal, Fundus Oculi, Intravitreal Injections, Male, Rabbits, Retina pathology, Retinal Diseases chemically induced, Retinal Diseases metabolism, Vascular Endothelial Growth Factor A administration & dosage, Blood-Retinal Barrier drug effects, Fluorescein Angiography methods, Retina drug effects, Retinal Diseases diagnosis, Vascular Endothelial Growth Factor A toxicity
- Abstract
Purpose: To determine the utility of polychromatic angiography (PCA) in the assessment of VEGF-induced blood retinal barrier (BRB) dysfunction in rabbits., Methods: Twenty-six eyes of 24 Dutch Belted rabbits were injected intravitreally with 1.25 μg (group A, n = 5), 10 μg (group C, n = 7), or 4 μg (group B, n = 6; group D, n = 4; and group E, n = 4) of VEGF on day 0. Groups D and E were also injected intravitreally with 1.25 μg and 12.5 μg bevacizumab, respectively, on day 2. On days 0, 2, 4, 7, 11, and 14, PCA was performed using a contrast agent mixture composed of fluorescein sodium, indocyanine green, PCM102, and PCM107 and imaged with a modified fundus camera. PCA scores were based on detected leaking fluorophores., Results: On day 7, there was a statistically significant difference between PCA scores of group A (0.6 ± 0.89) and both groups B (2.67 ± 1.37, P = 0.0154) and C (3.33 ± 0.52, P = 0.00085). There was also a statistically significant difference between groups B and E (PCA score 0.75 ± 0.96, P = 0.032) on day 7. On day 11, there was statistically significant difference between group C (1.80 ± 1.1) and both groups A (0, P = 0.021) and B (0.33 ± 0.52, P = 0.037)., Conclusions: A differential response to both increasing VEGF dose and administration of bevacizumab could be discerned using the PCA. PCA allowed stratification of VEGF-induced BRB dysfunction and inhibitory effects of bevacizumab therapy in the rabbit retina.
- Published
- 2013
- Full Text
- View/download PDF
50. Glucocorticoid induction of occludin expression and endothelial barrier requires transcription factor p54 NONO.
- Author
-
Keil JM, Liu X, and Antonetti DA
- Subjects
- Animals, Blood-Retinal Barrier metabolism, Blotting, Western, Cattle, Cells, Cultured, Chromatin Immunoprecipitation, Claudin-5 metabolism, DNA-Binding Proteins, Electrophoretic Mobility Shift Assay, Endothelium, Vascular metabolism, Gene Silencing physiology, Humans, Male, Nuclear Matrix-Associated Proteins genetics, Octamer Transcription Factors genetics, PTB-Associated Splicing Factor, RNA, Small Interfering genetics, RNA-Binding Proteins genetics, Rats, Retinal Vessels cytology, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Tandem Mass Spectrometry, Tight Junctions drug effects, Trans-Activators, Transfection, Blood-Retinal Barrier drug effects, Dexamethasone pharmacology, Endothelium, Vascular drug effects, Glucocorticoids pharmacology, Nuclear Matrix-Associated Proteins metabolism, Occludin biosynthesis, Octamer Transcription Factors metabolism, RNA-Binding Proteins metabolism
- Abstract
Purpose: Glucocorticoids (GCs) effectively reduce retinal edema and induce vascular barrier properties but possess unwanted side effects. Understanding GC induction of barrier properties may lead to more effective and specific therapies. Previous work identified the occludin enhancer element (OEE) as a GC-responsive cis-element in the promoters of multiple junctional genes, including occludin, claudin-5, and cadherin-9. Here, we identify two OEE-binding factors and determine their contribution to GC induction of tight junction (TJ) gene expression and endothelial barrier properties., Methods: OEE-binding factors were isolated from human retinal endothelial cells (HREC) using DNA affinity purification followed by MALDI-TOF MS/MS. Chromatin immunoprecipitation (ChIP) assays determined in situ binding. siRNA was used to evaluate the role of trans-acting factors in transcription of TJ genes in response to GC stimulation. Paracellular permeability was determined by quantifying flux through a cell monolayer, whereas transendothelial electrical resistance (TER) was measured using the ECIS system., Results: MS/MS analysis of HREC nuclear extracts identified the heterodimer of transcription factors p54/NONO (p54) and polypyrimidine tract-binding protein-associated splicing factor (PSF) as OEE-binding factors, which was confirmed by ChIP assay from GC-treated endothelial cells and rat retina. siRNA knockdown of p54 demonstrated that this factor is necessary for GC induction of occludin and claudin-5 expression. Further, p54 knockdown ablated the pro-barrier effects of GC treatment., Conclusions: p54 is essential for GC-mediated expression of occludin, claudin-5, and barrier induction, and the p54/PSF heterodimer may contribute to normal blood-retinal barrier (BRB) induction in vivo. Understanding the mechanism of GC induction of BRB properties may provide novel therapies for macular edema.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.