Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 24(12)2023 Jun 17.
Article in English | MEDLINE | ID: mdl-37373423

ABSTRACT

Accumulation of 7-ketocholesterol (7KC) occurs in age-related macular degeneration (AMD) and was found previously to promote fibrosis, an untreatable cause of vision loss, partly through induction of endothelial-mesenchymal transition. To address the hypothesis that 7KC causes mesenchymal transition of retinal pigment epithelial cells (RPE), we exposed human primary RPE (hRPE) to 7KC or a control. 7KC-treated hRPE did not manifest increased mesenchymal markers, but instead maintained RPE-specific proteins and exhibited signs of senescence with increased serine phosphorylation of histone H3, serine/threonine phosphorylation of mammalian target of rapamycin (p-mTOR), p16 and p21, ß-galactosidase labeling, and reduced LaminB1, suggesting senescence. The cells also developed senescence-associated secretory phenotype (SASP) determined by increased IL-1ß, IL-6, and VEGF through mTOR-mediated NF-κB signaling, and reduced barrier integrity that was restored by the mTOR inhibitor, rapamycin. 7KC-induced p21, VEGF, and IL-1ß were inhibited by an inhibitor of protein kinase C. The kinase regulates IQGAP1 serine phosphorylation. Furthermore, after 7KC injection and laser-induced injury, mice with an IQGAP1 serine 1441-point mutation had significantly reduced fibrosis compared to littermate control mice. Our results provide evidence that age-related accumulation of 7KC in drusen mediates senescence and SASP in RPE, and IQGAP1 serine phosphorylation is important in causing fibrosis in AMD.


Subject(s)
Choroidal Neovascularization , Macular Degeneration , Animals , Humans , Mice , Cellular Senescence , Choroidal Neovascularization/pathology , Fibrosis , Macular Degeneration/metabolism , Mammals/metabolism , Phosphorylation , Retinal Pigment Epithelium/metabolism , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , ras GTPase-Activating Proteins/metabolism
2.
Pediatr Res ; 94(3): 950-955, 2023 09.
Article in English | MEDLINE | ID: mdl-37016003

ABSTRACT

BACKGROUND: Recent clinical studies suggest that preeclampsia, characterized by uteroplacental insufficiency (UPI) and infant intrauterine growth restriction (IUGR), may be protective against retinopathy of prematurity (ROP) in preterm infants. Experimental models of UPI/IUGR have found an association of erythropoietin (EPO) with less severe oxygen-induced retinopathy (OIR); however, it is unclear if EPO/EPO receptor (EPOR) signaling was involved. We hypothesized that maternal UPI and resultant infant IUGR would protect against features of ROP through EPO/EPOR signaling. METHODS: We compared transgenic mice with hypoactive EPOR signaling (hWtEPOR) to littermate wild-type mice (mWtEpoR) in a novel combined model of IUGR and ROP. Thromboxane A2 (TXA2) was infused into pregnant C57Bl/6J dams to produce UPI/IUGR; postnatal pups and their foster dams were subjected to a murine OIR model. RESULTS: Following hyperoxia, hematocrits were similar between littermate wild-type (mWtEpoR) TXA2/OIR and vehicle/OIR pups. mWtEpoR TXA2/OIR had increased serum EPO, retinal EPO and VEGF, and decreased avascular retinal area (AVA) compared to vehicle/OIR pups. In comparison to the mWtEpoR TXA2/OIR pups, AVA was not reduced in hWtEPOR TXA2/OIR pups. CONCLUSION: Our findings provide biologic evidence that UPI/OIR-induced endogenous EPOR signaling confers protection against hyperoxia-induced vascular damage that may be related to pathophysiology in ROP. IMPACT: Maternal preeclampsia and infant growth restriction confer retinovascular protection against high oxygen-induced damage through endogenous erythropoietin signaling.


Subject(s)
Erythropoietin , Hyperoxia , Pre-Eclampsia , Retinal Neovascularization , Retinopathy of Prematurity , Infant, Newborn , Pregnancy , Humans , Female , Animals , Mice , Retinopathy of Prematurity/prevention & control , Retinopathy of Prematurity/complications , Hyperoxia/complications , Disease Models, Animal , Infant, Premature , Oxygen , Mice, Transgenic , Animals, Newborn , Mice, Inbred C57BL
3.
Biomedicines ; 10(7)2022 Jul 09.
Article in English | MEDLINE | ID: mdl-35884958

ABSTRACT

Erythropoietin (EPO) has been proposed to reduce the progression of atrophic age-related macular degeneration (AMD) due to its potential role in neuroprotection. However, overactive EPO receptor (EPOR) signaling increased laser-induced choroidal neovascularization (CNV) and choroidal macrophage number in non-lasered mice, which raised the question of whether EPOR signaling increased CNV through the recruitment of macrophages to the choroid that released pro-angiogenic factors or through direct angiogenic effects on endothelial cells. In this study, we addressed the hypothesis that EPOR signaling increased CNV by direct effects on macrophages or endothelial cells. We used tamoxifen-inducible macrophage-specific or endothelial cell-specific EPOR knockout mice in the laser-induced CNV model, and cultured choroidal endothelial cells isolated from adult human donors. We found that macrophage-specific knockout of EPOR influenced laser-induced CNV in females only, whereas endothelial-specific knockout of EPOR reduced laser-induced CNV in male mice only. In cultured human choroidal endothelial cells, knockdown of EPOR reduced EPO-induced signal transducer and activator of transcription 3 (STAT3) activation. Taken together, our findings suggest that EPOR signaling in macrophages or choroidal endothelial cells regulates the development of CNV in a sex-dependent manner. Further studies regarding the role of EPO-induced signaling are required to assess EPO safety and to select or develop appropriate therapeutic approaches.

4.
Int J Mol Sci ; 23(13)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35806359

ABSTRACT

The pathophysiology of retinopathy of prematurity (ROP) is postulated to first involve delayed intraretinal vascularization, followed by intravitreal neovascularization (IVNV). Although intravitreal agents that reduce the bioactivity of vascular endothelial growth factor (VEGF) are used to treat IVNV, concerns exist regarding their effects on intraretinal vascularization. In an experimental ROP model, VEGF receptor 2 (VEGFR2) knockdown in retinal endothelial cells reduced IVNV and promoted intraretinal vascularization, whereas knockdown of a downstream effector, signal transducer and activator of transcription 3 (STAT3) in retinal endothelial cells only reduced IVNV. In this study, we tested the hypothesis that the different pathways involved in VEGF-triggered VEGFR2 signaling and VEGF-triggered STAT3 signaling in retinal endothelial cells would allow us to delineate signaling pathways involved in IVNV from those involved in intraretinal vascularization in ROP. To address our hypothesis, we used RNA-sequencing and pathway enrichment analysis to determine changes in the transcriptome of cultured human retinal microvascular endothelial cells (HRMECs). Of the enriched pathways, inactivation of oncostatin M signaling was predicted by either KDR or STAT3 knockdown in the presence of VEGF. Activation of kinetochore metaphase signaling was predicted by KDR knockdown, whereas inactivation was predicted by STAT3 knockdown in the presence of VEGF. Inactivation of signaling by the Rho family of GTPases was predicted by KDR knockdown, but activation was predicted by STAT3 knockdown in the presence of VEGF. Taken together, our data identified unique signaling pathway differences between VEGF-triggered VEGFR2 and VEGF-triggered STAT3 in HRMECs that might have implications in ROP.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Infant, Newborn , Neovascularization, Pathologic/metabolism , RNA-Seq , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
5.
Front Pediatr ; 9: 796143, 2021.
Article in English | MEDLINE | ID: mdl-34956992

ABSTRACT

Retinopathy of prematurity (ROP) is a leading cause of blindness in children worldwide. Blindness can occur from retinal detachment caused by pathologic retinal angiogenesis into the vitreous, termed intravitreal neovascularization (IVNV). Although agents that interfere with the bioactivity of vascular endothelial growth factor (VEGF) are now used to treat IVNV, concerns exist regarding the identification of optimal doses of anti-VEGF for individual infants and the effect of broad VEGF inhibition on physiologic angiogenesis in external organs or in the retina of a preterm infant. Therefore, it is important to understand VEGF signaling in both physiologic and pathologic angiogenesis in the retina. In this manuscript, we review the role of receptors that interact with VEGF in oxygen-induced retinopathy (OIR) models that represent features of ROP pathology. Specifically, we discuss our work regarding the regulation of VEGFR2 signaling in retinal endothelial cells to not only reduce severe ROP but also facilitate physiologic retinal vascular and neuronal development.

6.
Clin Ophthalmol ; 15: 4475-4484, 2021.
Article in English | MEDLINE | ID: mdl-34819719

ABSTRACT

PURPOSE: We assessed the effect of two lens cube sizes, three tip sizes, and two ultrasound (US) approaches on phacoemulsification efficiency and chatter. METHODS: After porcine lens nuclei were soaked in formalin, we divided them into cubes measuring 2.0 mm or 3.0 mm. We collected efficiency and chatter data for 30-degree bent 19 G, 20 G, and 21 G tips with a continuous torsional US system; and for straight 19 G, 20 G, and 21 G tips with a micropulse longitudinal US system. RESULTS: The average time needed for removal was always higher for the 3.0 mm lens cube than for the 2.0 mm lens cube. Statistically significant differences were observed between the 19 G and 21 G tips with micropulse longitudinal US using a 2.0 mm cube and a 3.0 mm cube, and with continuous transversal US using a 2.0 mm lens cube and a 3.0 mm cube. We did not observe significant differences between 19 G and 20 G tips with either cube size in either US system. However, we noted identical trends for both cube sizes with both US approaches; 19 G tips performed better than 20 G and 21 G tips. CONCLUSION: Regardless of the lens size, 19 G needles were the most efficient, and had both the fewest outliers and the smallest standard deviations.

7.
Cells ; 10(9)2021 09 14.
Article in English | MEDLINE | ID: mdl-34572063

ABSTRACT

Age-related macular degeneration (AMD) is one of the leading causes of blindness worldwide. Vision loss from the neovascular form is associated with the invasion of choroidal endothelial cells into the neural retina to form vision-threatening macular neovascularization (MNV). Anti-angiogenic agents are the current standard of care but are effective in only ~50% of AMD cases. The molecular mechanisms involved in invasive MNV point to the importance of regulating signaling pathways that lead to pathologic biologic outcomes. In studies testing the effects of AMD-related stresses, activation of the Rho GTPase, Rac1, was found to be important for the choroidal endothelial cell invasion into the neural retina. However, current approaches to prevent Rac1 activation are inefficient and less effective. We summarize active Rac1-mediated mechanisms that regulate choroidal endothelial cell migration. Specifically, we discuss our work regarding the role of a multidomain protein, IQ motif containing GTPase activating protein 1 (IQGAP1), in sustaining pathologic Rac1 activation and a mechanism by which active Rap1, a Ras-like GTPase, may prevent active Rac1-mediated choroidal endothelial cell migration.


Subject(s)
Cell Movement , Choroid/pathology , Endothelial Cells/pathology , Macular Degeneration/pathology , Neovascularization, Pathologic/pathology , rac1 GTP-Binding Protein/metabolism , Animals , Choroid/metabolism , Endothelial Cells/metabolism , Humans , Macular Degeneration/etiology , Macular Degeneration/metabolism , Neovascularization, Pathologic/metabolism
8.
FASEB J ; 35(7): e21642, 2021 07.
Article in English | MEDLINE | ID: mdl-34166557

ABSTRACT

Neovascular age-related macular degeneration (nAMD) is a leading cause of blindness. The pathophysiology involves activation of choroidal endothelial cells (CECs) to transmigrate the retinal pigment epithelial (RPE) monolayer and form choroidal neovascularization (CNV) in the neural retina. The multidomain GTPase binding protein, IQGAP1, binds active Rac1 and sustains activation of CECs, thereby enabling migration associated with vision-threatening CNV. IQGAP1 also binds the GTPase, Rap1, which when activated reduces Rac1 activation in CECs and CNV. In this study, we tested the hypothesis that active Rap1 binding to IQGAP1 is necessary and sufficient to reduce Rac1 activation in CECs, and CNV. We found that pharmacologic activation of Rap1 or adenoviral transduction of constitutively active Rap1a reduced VEGF-mediated Rac1 activation, migration, and tube formation in CECs. Following pharmacologic activation of Rap1, VEGF-mediated Rac1 activation was reduced in CECs transfected with an IQGAP1 construct that increased active Rap1-IQGAP1 binding but not in CECs transfected with an IQGAP1 construct lacking the Rap1 binding domain. Specific knockout of IQGAP1 in endothelial cells reduced laser-induced CNV and Rac1 activation in CNV lesions, but pharmacologic activation of Rap1 did not further reduce CNV compared to littermate controls. Taken together, our findings provide evidence that active Rap1 binding to the IQ domain of IQGAP1 is sufficient to interfere with active Rac1-mediated CEC activation and CNV formation.


Subject(s)
Choroid/metabolism , Choroidal Neovascularization/prevention & control , Endothelial Cells/metabolism , Protein Interaction Domains and Motifs , rap1 GTP-Binding Proteins/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , Cell Movement , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Female , Male , Mice , Mice, Inbred C57BL , Signal Transduction , rap1 GTP-Binding Proteins/genetics , ras GTPase-Activating Proteins/genetics
9.
Angiogenesis ; 24(3): 583-595, 2021 08.
Article in English | MEDLINE | ID: mdl-33646466

ABSTRACT

Oxidized cholesterols and lipids accumulate in Bruch's membrane in age-related macular degeneration (AMD). It remains unknown what causal relationship exists between these substances and AMD pathophysiology. We addressed the hypothesis that a prevalent form, 7-ketocholesterol (7KC), promotes choroidal endothelial cell (CEC) migration and macular neovascularization in AMD. Compared to control, 7KC injection caused 40% larger lectin-stained lesions, but 70% larger lesions measured by optical coherence tomography one week after laser-injury. At two weeks, 7KC-injected eyes had 86% larger alpha smooth muscle actin (αSMA)-labeled lesions and more collagen-labeling than control. There was no difference in cell death. 7KC-treated RPE/choroids had increased αSMA but decreased VE-cadherin. Compared to control-treated CECs, 7KC unexpectedly reduced endothelial VE-cadherin, CD31 and VEGFR2 and increased αSMA, fibroblast activation protein (FAP) and transforming growth factor beta (TGFß). Inhibition of TGFß receptor-mediated signaling by SB431542 abrogated 7KC-induced loss of endothelial and increase in mesenchymal proteins in association with decreased transcription factor, SMAD3. Knockdown of SMAD3 partially inhibited 7KC-mediated loss of endothelial proteins and increase in αSMA and FAP. Compared to control, 7KC-treatment of CECs increased Rac1GTP and migration, and both were inhibited by the Rac1 inhibitor; however, CECs treated with 7KC had reduced tube formation. These findings suggest that 7KC, which increases in AMD and with age, induces mesenchymal transition in CECs making them invasive and migratory, and causing fibrosis in macular neovascularization. Further studies to interfere with this process may reduce fibrosis and improve responsiveness to anti-VEGF treatment in non-responsive macular neovascularization in AMD.


Subject(s)
Cell Movement/drug effects , Choroid , Choroidal Neovascularization , Endothelial Cells , Ketocholesterols/adverse effects , Macular Degeneration , Animals , Choroid/blood supply , Choroid/metabolism , Choroid/pathology , Choroidal Neovascularization/chemically induced , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Fibrosis , Ketocholesterols/pharmacology , Macular Degeneration/chemically induced , Macular Degeneration/metabolism , Macular Degeneration/pathology , Mice
10.
Invest Ophthalmol Vis Sci ; 61(10): 23, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32785675

ABSTRACT

Purpose: Exogenous erythropoietin (EPO) is being considered for tissue protection and angiogenesis in retinal vascular diseases. However, studies are limited by insufficient tools to address signaling effects through the EPO receptor (EPOR). We used a humanized mouse model of hypoactive EPOR signaling to test the hypothesis that EPOR signaling supports angiogenesis in retinovascular diseases. Methods: Humanized Knockin EPOR mice (hWtEPOR) with hypoactive EPOR signaling were compared to littermate wild-type mice (WT). Postnatal day (p)7 mice of each genotype were exposed to 75% oxygen for five days, followed by 21% oxygen in the oxygen-induced retinopathy model (OIR) and compared to room-air (RA)-raised pups. At time points after OIR, pups were sacrificed, and flat-mounted, lectin-stained retinas were analyzed for central avascular area or intravitreal neovascular area (IVNV). Flash-frozen retinas were analyzed for angiogenic protein (Epo, VEGF, p-Stat3) and gene (Vegfa, Kdr, Epo, Hif1α, Hif2α) expression levels. Results: In OIR, hWtEPOR mice had increased AVA compared with WT at p8, p12, and p17, but there was no difference in IVNV between hWtEPOR and WT mice at p17. Although VEGF and p-STAT3 proteins were increased in WT at p17 OIR, there were no differences in retinal angiogenic factor expression levels between hWtEPOR and WT OIR at p17 despite similar areas of IVNV. Conclusions: Our data support the hypothesis that EPOR signaling was associated with regrowth of vascularization following oxygen-induced capillary dropout and played a role in intravitreal angiogenesis. Additional study of EPOR signaling regulation on other angiogenic factor pathways may be considered.


Subject(s)
Neovascularization, Pathologic/metabolism , Receptors, Erythropoietin/metabolism , Retinal Neovascularization/metabolism , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Erythropoietin/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen , Real-Time Polymerase Chain Reaction , Retina/pathology , Retinal Neovascularization/pathology , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
11.
Angiogenesis ; 23(4): 685-698, 2020 11.
Article in English | MEDLINE | ID: mdl-32783108

ABSTRACT

Loss of visual acuity in neovascular age-related macular degeneration (nAMD) occurs when factors activate choroidal endothelial cells (CECs) to transmigrate the retinal pigment epithelium into the sensory retina and develop into choroidal neovascularization (CNV). Active Rac1 (Rac1GTP) is required for CEC migration and is induced by different AMD-related stresses, including vascular endothelial growth factor (VEGF). Besides its role in pathologic events, Rac1 also plays a role in physiologic functions. Therefore, we were interested in a method to inhibit pathologic activation of Rac1. We addressed the hypothesis that IQGAP1, a scaffold protein with a Rac1 binding domain, regulates pathologic Rac1GTP in CEC migration and CNV. Compared to littermate Iqgap1+/+, Iqgap1-/- mice had reduced volumes of laser-induced CNV and decreased Rac1GTP and phosphorylated VEGFR2 (p-VEGFR2) within lectin-stained CNV. Knockdown of IQGAP1 in CECs significantly reduced VEGF-induced Rac1GTP, mediated through p-VEGFR2, which was necessary for CEC migration. Moreover, sustained activation of Rac1GTP induced by VEGF was eliminated when CECs were transfected with an IQGAP1 construct that is unable to bind Rac1. IQGAP1-mediated Src activation was involved in initiating Rac1 activation, CEC migration, and tube formation. Our findings indicate that CEC IQGAP1 interacts with VEGFR2 to mediate Src activation and subsequent Rac1 activation and CEC migration. In addition, IQGAP1 binding to Rac1GTP results in sustained activation of Rac1, leading to CEC migration toward VEGF. Our study supports a role of IQGAP1 and the interaction between IQGAP1 and Rac1GTP to restore CECs quiescence and, therefore, prevent vision-threatening CNV in nAMD.


Subject(s)
Choroidal Neovascularization/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , rac1 GTP-Binding Protein/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , Cell Movement , Choroid/pathology , Choroidal Neovascularization/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Enzyme Activation , Humans , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Protein Domains , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , ras GTPase-Activating Proteins/chemistry , src-Family Kinases/metabolism
12.
Am J Pathol ; 190(3): 630-641, 2020 03.
Article in English | MEDLINE | ID: mdl-32093902

ABSTRACT

The investigation of erythropoietin (EPO) has expanded to include potential nonhematopoietic roles in neural and retinal diseases, including diabetic retinopathy. However, it remains unclear how EPO functions to support the neural retina. Transgenic mice with hypoactive EPO receptor (EPOR) signaling (hWtEPOR) were compared with littermate control mice (WT) to test the role of EPOR signaling under normal conditions and after vascular injury and regrowth into the retina. Although retinal function tested with OptoMotry and electroretinography was comparable to adult (8-week-old) littermate WT mice, hWtEPOR mice had thinner inner and outer plexiform layers and a greater number of amacrine cells. Injury and repair caused by the oxygen-induced retinopathy model reduced visual acuity thresholds, reduced electroretinography amplitudes, and thinned the outer plexiform and inner nuclear layers of both WT and hWtEPOR 8-week-old mice. In hWtEPOR compared with WT mice, scotopic a-wave amplitudes were reduced by injury, despite no change in outer nuclear layer thickness; and peripheral rod, but not cone number, was reduced. Scotopic b-waves were reduced in injured hWtEPOR mice compared with WT, and rod bipolar cell ectopic neurites were increased in both genotypes after injury, suggesting a potential reparative process to preserve connectivity and the b-wave. Normal EPOR signaling appeared important because ectopic neurites and b-waves were lower in the hWtEPOR than WT injured mice.


Subject(s)
Diabetic Retinopathy/physiopathology , Erythropoietin/metabolism , Receptors, Erythropoietin/metabolism , Retinal Diseases/physiopathology , Signal Transduction , Vascular System Injuries/physiopathology , Animals , Electroretinography , Erythropoietin/genetics , Female , Male , Mice , Mice, Transgenic , Receptors, Erythropoietin/genetics , Retina/physiopathology
13.
Mol Cell Biol ; 36(15): 2051-66, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27185880

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.


Subject(s)
Epithelial Cells/cytology , G-Protein-Coupled Receptor Kinases/metabolism , Occludin/metabolism , Serine/metabolism , Tight Junctions/metabolism , Animals , Cell Proliferation , Dogs , Epithelial Cells/metabolism , Humans , Madin Darby Canine Kidney Cells , Occludin/chemistry , Occludin/genetics , Phosphorylation , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...