Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neuroinflammation ; 20(1): 238, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37858232

ABSTRACT

BACKGROUND: Neovascular age-related macular degeneration causes vision loss from destructive angiogenesis, termed choroidal neovascularization (CNV). Cx3cr1-/- mice display alterations in non-classical monocytes and microglia with increased CNV size, suggesting that non-classical monocytes may inhibit CNV formation. NR4A1 is a transcription factor that is necessary for maturation of non-classical monocytes from classical monocytes. While Nr4a1-/- mice are deficient in non-classical monocytes, results are confounded by macrophage hyper-activation. Nr4a1se2/se2 mice lack a transcriptional activator, resulting in non-classical monocyte loss without macrophage hyper-activation. MAIN BODY: We subjected Nr4a1-/- and Nr4a1se2/se2 mice to the laser-induced CNV model and performed multi-parameter flow cytometry. We found that both models lack non-classical monocytes, but only Nr4a1-/- mice displayed increased CNV area. Additionally, CD11c+ macrophages were increased in Nr4a1-/- mice. Single-cell transcriptomic analysis uncovered that CD11c+ macrophages were enriched from Nr4a1-/- mice and expressed a pro-angiogenic transcriptomic profile that was disparate from prior reports of macrophage hyper-activation. CONCLUSIONS: These results suggest that non-classical monocytes are dispensable during CNV, and NR4A1 deficiency results in increased recruitment of pro-angiogenic macrophages.


Subject(s)
Choroidal Neovascularization , Macular Degeneration , Animals , Mice , Choroidal Neovascularization/genetics , Disease Models, Animal , Macrophages/physiology , Macular Degeneration/genetics , Mice, Inbred C57BL , Microglia , Monocytes
2.
Exp Eye Res ; 237: 109703, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38652673

ABSTRACT

We previously showed that macrophage-like cells (MLCs) are increased in eyes with advanced diabetic retinopathy (DR). Here, we hypothesized that MLC density was correlated with ischemia using optical coherence tomography angiography (OCTA) and ultra-widefield fluorescein angiography (UWF-FA). Treatment-naïve diabetic eyes were prospectively imaged with repeated OCTA (average 5.3 scans per eye) and UWF-FA imaging. OCTA images were registered and averaged to generate a superficial capillary plexus (SCP), deep capillary plexus (DCP), and MLC slab. We calculated geometric perfusion deficit (GPD), vessel length density, and vessel density for the SCP and DCP. MLC density was quantified by two masked graders and averaged. Ischemia on UWF-FA was measured to generate a non-perfusion area (NPA) and index (NPI). Since MLC density was non-parametrically distributed, MLC density was correlated with ischemia metrics using Spearman correlations. Forty-five treatment-naïve eyes of 45 patients (59 ± 12 years of age; 56% female) were imaged. We included 6 eyes with no DR, 7 eyes with mild non-proliferative DR (NPDR), 22 moderate NPDR, 4 severe NPDR, and 6 PDR eyes. MLC density between graders was highly correlated (r = 0.9592, p < 0.0001). MLC density was correlated with DCP GPD (r = 0.296, p = 0.049), but no other OCTA ischemia metrics. MLC density was also correlated with UWF-FA NPA (r = 0.330, p = 0.035) and NPI (r = 0.332, p = 0.034). MLC density was correlated with total ischemia on UWF-FA and local DCP GPD. Since both UWF-FA and DCP non-perfusion are associated with higher risk for DR progression, MLC density could be another potential biomarker for DR progression.


Subject(s)
Diabetic Retinopathy , Fluorescein Angiography , Ischemia , Macrophages , Retinal Vessels , Tomography, Optical Coherence , Humans , Female , Middle Aged , Male , Tomography, Optical Coherence/methods , Fluorescein Angiography/methods , Diabetic Retinopathy/physiopathology , Diabetic Retinopathy/pathology , Retinal Vessels/pathology , Retinal Vessels/physiopathology , Retinal Vessels/diagnostic imaging , Prospective Studies , Cell Count , Ischemia/physiopathology , Ischemia/pathology , Macrophages/pathology , Aged , Fundus Oculi
3.
Arterioscler Thromb Vasc Biol ; 42(11): 1413-1427, 2022 11.
Article in English | MEDLINE | ID: mdl-36172864

ABSTRACT

BACKGROUND: The choroidal vasculature, including the choriocapillaris and vortex veins, is essential for providing nutrients to the metabolically demanding photoreceptors and retinal pigment epithelium. Choroidal vascular dysfunction leads to vision loss and is associated with age-related macular degeneration and the poorly understood pachychoroid diseases including central serous chorioretinopathy and polypoidal choroidal vasculopathy that are characterized by formation of dilated pachyvessels throughout the choroid. METHODS: Using neural crest-specific Angpt1 knockout mice, we show that Angiopoietin 1, a ligand of the endothelial receptor TEK (also known as Tie2) is essential for choriocapillaris development and vortex vein patterning. RESULTS: Lacking choroidal ANGPT1, neural crest-specific Angpt1 knockout eyes exhibited marked choriocapillaris attenuation and 50% reduction in number of vortex veins, with only 2 vortex veins present in the majority of eyes. Shortly after birth, dilated choroidal vessels resembling human pachyvessels were observed extending from the remaining vortex veins and displacing the choriocapillaris, leading to retinal pigment epithelium dysfunction and subretinal neovascularization similar to that seen in pachychoroid disease. CONCLUSIONS: Together, these findings identify a new role for ANGPT1 in ocular vascular development and demonstrate a clear link between vortex vein dysfunction, pachyvessel formation, and disease.


Subject(s)
Angiopoietin-1 , Central Serous Chorioretinopathy , Humans , Mice , Animals , Angiopoietin-1/genetics , Ligands , Tomography, Optical Coherence , Choroid/blood supply , Retrospective Studies
4.
J Neuroophthalmol ; 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37585271

ABSTRACT

BACKGROUND: Cerebral vascular malformations (CVMs) may result in hemorrhage, seizure, neurologic dysfunction, and death. CVMs include capillary telangiectasias, venous malformations, cavernous malformations, and arteriovenous malformations. Cavernous and arteriovenous malformations carry the highest risk of complications. Retinal venous malformations (RVMs) have been proposed as an associated finding. Our objective was to determine the prevalence of RVMs in patients with high-risk CVMs. METHODS: We retrospectively reviewed patients diagnosed with cerebral cavernous or arteriovenous malformations (high-risk CVMs) who were evaluated by the ophthalmology service at Northwestern University between 2017 and 2020. Patients were stratified into 3 cohorts based on level of certainty: dilated funduscopic examination (DFE), DFE with any form of ocular imaging, and DFE with complete imaging of the macula. We recorded ophthalmic examination abnormalities, ocular imaging findings, and major CVM complications. RESULTS: We evaluated 156 patients with high-risk CVMs who had undergone DFE. Ocular imaging of any type was performed in 56 patients, of whom 46 had complete imaging of the macula. Zero RVMs were identified in any cohort (95% confidence interval: 0%-1.9% for the entire cohort, 0%-5.4% for any ocular imaging cohort, and 0%-6.5% for the complete macular imaging cohort). Cerebral hemorrhage or seizure occurred in 15%-33% of patients. Associated visual field defects or cranial nerve palsies were found in 14%-20% of patients. CONCLUSIONS: Zero RVMs were identified in patients with high-risk CVMs. However, neuro-ophthalmic findings were common. Therefore, we recommend neuroimaging for patients with RVMs and neuro-ophthalmic signs or symptoms. In asymptomatic patients with RVMs, a potential algorithm for neuroimaging is proposed.

5.
J Neuroinflammation ; 19(1): 203, 2022 Aug 08.
Article in English | MEDLINE | ID: mdl-35941655

ABSTRACT

BACKGROUND: Diabetic retinopathy and retinal vein occlusion are vision threatening retinal vascular diseases. Current first-line therapy targets the vascular component, but many patients are treatment-resistant due to unchecked inflammation. Non-invasive inflammatory imaging biomarkers are a significant unmet clinical need for patients. Imaging of macrophage-like cells on the surface of the retina using clinical optical coherence tomography (OCT) is an emerging field. These cells are increased in patients with retinal vascular disease, and could be a potential inflammatory biomarker. However, since OCT is limited by an axial resolution of 5-10 microns, the exact location and identity of these retinal cells is currently unknown. METHODS: We performed OCT followed by confocal immunofluorescence in wild-type mice to identify macrophages within 5-10 microns of the vitreoretinal interface. Next, we used Cx3cr1CreER/+; Rosa26zsGreen/+ mice to fate map retinal surface macrophages. Using confocal immunofluorescence of retinal sections and flatmounts, we quantified IBA1+Tmem119+CD169neg microglia, IBA1+Tmem119negCD169neg perivascular macrophages, and IBA1+Tmem119negCD169+ vitreal hyalocytes. Finally, we modeled neuroinflammation with CCL2 treatment and characterized retinal surface macrophages using flow cytometry, OCT, and confocal immunofluorescence. RESULTS: We were able to detect IBA1+ macrophages within 5-10 microns of the vitreoretinal interface in wild-type mice using OCT followed by confirmatory confocal immunofluorescence. Retinal surface macrophages were 83.5% GFP+ at Week 1 and 82.4% GFP+ at Week 4 using fate mapping mice. At steady state, these macrophages included 82% IBA1+Tmem119+CD169neg microglia, 9% IBA1+Tmem119negCD169+ vitreal hyalocytes, and 9% IBA1+Tmem119negCD169neg perivascular macrophages. After CCL2-driven neuroinflammation, many Ly6C+ cells were detectable on the retinal surface using OCT followed by confocal immunofluorescence. CONCLUSIONS: Macrophages within close proximity to the vitreoretinal interface are self-renewing cells, and predominantly microglia with minor populations of perivascular macrophages and vitreal hyalocytes at steady state. In the context of neuroinflammation, monocytes and monocyte-derived macrophages are a significant component of retinal surface macrophages. Human OCT-based imaging of retinal surface macrophages is a potential biomarker for inflammation during retinal vascular disease.


Subject(s)
Retinal Diseases , Retinal Vein Occlusion , Animals , Biomarkers , Disease Models, Animal , Humans , Inflammation/diagnostic imaging , Macrophages , Mice , Microglia
6.
J Neuroinflammation ; 17(1): 341, 2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33187533

ABSTRACT

BACKGROUND: Neovascular age-related macular degeneration (nAMD) commonly causes vision loss from aberrant angiogenesis, termed choroidal neovascularization (CNV). Macrophages are heterogeneous cells that are necessary for experimental CNV, present in human CNV samples, and can display diverse functions, which are dependent upon both their origin and tissue microenvironment. Despite these associations, choroidal macrophage heterogeneity remains unexplored. METHODS: We performed multi-parameter flow cytometry on wildtype (WT) and Ccr2-/- mice after laser injury to identify macrophage subtypes, and determine which subsets originate from classical monocytes. To fate map tissue resident macrophages at steady state and after laser injury, we used the Cx3cr1CreER/+ ; Rosa26zsGFP/+ mouse model. We reanalyzed previously published single-cell RNA-seq of human choroid samples from healthy and nAMD patients to investigate human macrophage heterogeneity, disease association, and function. RESULTS: We identified 4 macrophage subsets in mice: microglia, MHCII+CD11c-, MHCII+CD11c+, and MHCII-. Microglia are tissue resident macrophages at steady state and unaffected by laser injury. At steady state, MHCII- macrophages are long lived, tissue resident macrophages, while MHCII+CD11c- and MHCII+CD11c+ macrophages are partially replenished from blood monocytes. After laser injury, MHCII+CD11c- macrophages are entirely derived from classical monocytes, MHCII- macrophages originate from classical monocytes (90%) and an expansion of tissue resident macrophages (10%), and MHCII+CD11c+ macrophages are derived from classical monocytes (70%), non-classical monocytes (10%), and an expansion of tissue resident macrophages (20%). Single-cell RNA-seq analysis of human choroid found 5 macrophage subsets: two MHCII+CD11C- and three MHCII+CD11C+ populations. One MHCII+CD11C+ subset was 78% derived from a patient with nAMD. Differential expression analysis identified up-regulation of pro-angiogenic gene expression in one MHCII+CD11C- and two MHCII+CD11C+ subsets, including the disease-associated cluster. The upregulated MHCII+CD11C- pro-angiogenic genes were unique compared to the increased MHCII+CD11C+ angiogenesis genes. CONCLUSIONS: Macrophage origin impacts heterogeneity at steady state and after laser injury in mice. Both mice and human patients demonstrate similar macrophage subtypes. Two discrete pro-angiogenic macrophage populations exist in the human choroid. Targeting specific, pro-angiogenic macrophage subsets is a potential novel therapeutic for nAMD.


Subject(s)
Choroidal Neovascularization/genetics , Choroidal Neovascularization/metabolism , Genetic Heterogeneity , Macrophages/metabolism , Animals , Choroidal Neovascularization/pathology , Female , Laser Therapy/adverse effects , Macrophages/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
7.
Exp Eye Res ; 184: 30-37, 2019 07.
Article in English | MEDLINE | ID: mdl-30978346

ABSTRACT

A patient with bilateral diffuse uveal melanocytic proliferation (BDUMP) associated with endometrial cancer was treated with plasmapheresis, but failed therapy with progressive serous retinal detachment. We collected plasma before and after plasmapheresis therapy. Our goal was to determine if the cultured melanocyte elongation and proliferation (CMEP) factor and hepatocyte growth factor (HGF) was present in the IgG enriched fraction and understand why our patient failed plasmapheresis therapy. Melanocytes were cultured for 3-5 days in the presence of control medium, unfractionated pre-plasmapheresis BDUMP medium, IgG enriched or IgG depleted BDUMP medium, or unfractionated post-plasmapheresis BDUMP medium. Subretinal fluid was collected from patients with BDUMP and control retinal detachments and analyzed by electropheresis with immunoblotting. Medium with unfractionated BDUMP plasma stimulated melanocyte growth 1.4-1.5 fold compared to control medium on days 3-5 (p < 0.001 for all). Both IgG enriched and IgG depleted BDUMP medium mildly increased melanocyte growth 1.3 fold (p < 0.05 for enriched, p < 0.01 for depleted) compared to control. In comparison, unfractionated BDUMP medium caused a 1.7-fold increase in melanocyte growth, which was significantly more than the enriched (p < 0.01) and depleted (p < 0.05) fractions. Pre-plasmapheresis and post-plasmapheresis unfractionated BDUMP medium equally stimulated melanocyte growth 1.7-fold (p < 0.05) compared to control. HGF was present in IgG depleted, pre-plasmapheresis, and post-plasmapheresis samples, but absent in the IgG enriched fraction. There was no enrichment of IgG in the subretinal fluid from eyes with BDUMP. In conclusion, CMEP factor is not concentrated in the IgG enriched plasma fraction in our patient who failed plasmapheresis therapy. HGF levels have no correlation with melanocyte growth. Because plasmapheresis preferentially removes immunoglobulins from the plasma, our patient responded poorly to plasmapheresis treatment with worsening retinal detachment.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Endometrial Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/blood , Melanocytes/pathology , Paraneoplastic Syndromes, Ocular/pathology , Uvea/pathology , Adenocarcinoma, Clear Cell/blood , Adenocarcinoma, Clear Cell/therapy , Aged , Cell Proliferation , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Endometrial Neoplasms/blood , Endometrial Neoplasms/therapy , Female , Fluorescein Angiography , Humans , Immunoblotting , Multimodal Imaging , Paraneoplastic Syndromes, Ocular/blood , Paraneoplastic Syndromes, Ocular/therapy , Plasmapheresis , Subretinal Fluid , Treatment Failure
8.
Retina ; 39(10): 1861-1871, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30044267

ABSTRACT

PURPOSE: To determine the features of primary vitreoretinal lymphoma on multimodal ultra-widefield imaging and correlate these findings to clinical outcomes. METHODS: We report a retrospective, observational case series of 43 eyes of 23 patients with biopsy-proven B-cell primary vitreoretinal lymphoma. Fundus photography, fluorescein angiography (FA), optical coherence tomography, fundus autofluorescence, and indocyanine green angiography images were reviewed. Medical records were assessed for the central nervous system involvement and visual acuity outcomes at 6 and 12 months after presentation. RESULTS: Common fundus photography findings were sub-retinal pigment epithelium lesions and vitritis alone. Common ultra-widefield FA findings were vascular leakage and scleral staining. Retinal optical coherence tomography features overlying sub-retinal pigment epithelium lesions or within the macula predicted fluorescence patterns. The presence of retinal fluid or disorganization associated with hyperfluorescence and late leakage. Normal retinal structures associated with hypofluorescence of sub-retinal pigment epithelium lesions or macular leopard spotting on FA and fundus autofluorescence. Peripheral abnormalities noted on ultra-widefield fundus photography, FA, and indocyanine green angiography were more frequent than posterior pole abnormalities. No imaging characteristics predicted time to the central nervous system progression. CONCLUSION: Ultra-widefield imaging was more informative than posterior pole imaging in fundus photography, FA, and indocyanine green angiography. Common findings on multimodal ultra-widefield imaging may lead to early diagnostic vitrectomy and may reduce the delay in primary vitreoretinal lymphoma diagnosis.


Subject(s)
Fluorescein Angiography/methods , Multimodal Imaging/methods , Retinal Neoplasms/diagnosis , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence/methods , Vitreous Body/pathology , Adult , Aged , Aged, 80 and over , Choroid/pathology , Female , Follow-Up Studies , Fundus Oculi , Humans , Male , Middle Aged , Retrospective Studies , Visual Acuity
9.
Am J Physiol Endocrinol Metab ; 309(10): E819-28, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26394663

ABSTRACT

Cholecystokinin (CCK) is a peptide hormone produced in the gut and brain with beneficial effects on digestion, satiety, and insulin secretion. CCK is also expressed in pancreatic ß-cells, but only in models of obesity and insulin resistance. Whole body deletion of CCK in obese mice leads to reduced ß-cell mass expansion and increased apoptosis. We hypothesized that islet-derived CCK is important in protection from ß-cell apoptosis. To determine the specific role of ß-cell-derived CCK in ß-cell mass dynamics, we generated a transgenic mouse that expresses CCK in the ß-cell in the lean state (MIP-CCK). Although this transgene contains the human growth hormone minigene, we saw no expression of human growth hormone protein in transgenic islets. We examined the ability of MIP-CCK mice to maintain ß-cell mass when subjected to apoptotic stress, with advanced age, and after streptozotocin treatment. Aged MIP-CCK mice have increased ß-cell area. MIP-CCK mice are resistant to streptozotocin-induced diabetes and exhibit reduced ß-cell apoptosis. Directed CCK overexpression in cultured ß-cells also protects from cytokine-induced apoptosis. We have identified an important new paracrine/autocrine effect of CCK in protection of ß-cells from apoptotic stress. Understanding the role of ß-cell CCK adds to the emerging knowledge of classic gut peptides in intraislet signaling. CCK receptor agonists are being investigated as therapeutics for obesity and diabetes. While these agonists clearly have beneficial effects on body weight and insulin sensitivity in peripheral tissues, they may also directly protect ß-cells from apoptosis.


Subject(s)
Aging , Apoptosis , Cholecystokinin/metabolism , Diabetes Mellitus, Type 2/metabolism , Down-Regulation , Insulin-Secreting Cells/metabolism , Stress, Physiological , Animals , Cell Line , Cholecystokinin/genetics , Cytokines/adverse effects , Cytokines/genetics , Cytokines/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/prevention & control , Hyperglycemia/blood , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Insulin/genetics , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/pathology , Male , Mice, Transgenic , Promoter Regions, Genetic , Rats , Recombinant Proteins/adverse effects , Recombinant Proteins/metabolism , Streptozocin , Tissue Culture Techniques
10.
Invest Ophthalmol Vis Sci ; 65(4): 37, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38652648

ABSTRACT

Purpose: Adjuvant, pre-operative intravitreal anti-vascular endothelial growth factor (anti-VEGF) injections have been used to reduce peri-operative bleeding in eyes undergoing pars-plana vitrectomy for complications of proliferative diabetic retinopathy (PDR). To address the concern over their potential off-target effects of progressive fibrous contraction, we sought to dissect the transcriptional changes in the surgically extracted fibrovascular membranes (FVMs). Methods: We analyzed surgically extracted FVMs from 10 eyes: 4 eyes pretreated with intravitreal bevacizumab (IVB) and 6 untreated eyes. FVMs were digested into single cells, mRNA was extracted from endothelial cell-enriched (microbead selection with CD31) and non-endothelial cell compartments, followed by RT-qPCR quantification. We then compared the relative expression of genes involved in angiogenesis, endothelial cell integrity, and myofibroblastic processes between treated and untreated FVMs. Results: Endothelial cells from IVB pretreated FVMs showed significant reduction of VEGFA, VEGF receptors (FLT1 and KDR), and angiopoietin 2 expression as well as increased vascular endothelial cadherin and endothelin, suggesting reduced angiogenesis and enhanced vascular integrity. The non-endothelial cell fraction showed decreased expression of VEGFA and fibronectin, without significant difference in the expression of other profibrotic factors. Conclusions: Our findings confirm that adjuvant pre-operative IVB decreased fibronectin and increase endothelin-1 expression without affecting other profibrotic gene expression, uncovering an important interaction between IVB and endothelin-1 that deserves further study.


Subject(s)
Angiogenesis Inhibitors , Bevacizumab , Diabetic Retinopathy , Fibrosis , Intravitreal Injections , Vascular Endothelial Growth Factor A , Vitrectomy , Humans , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/surgery , Angiogenesis Inhibitors/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Bevacizumab/therapeutic use , Bevacizumab/pharmacology , Male , Female , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retinal Neovascularization/metabolism , Retinal Neovascularization/drug therapy , Aged , Preoperative Care , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology
11.
Front Immunol ; 15: 1399989, 2024.
Article in English | MEDLINE | ID: mdl-38799448

ABSTRACT

Introduction: Macrophage function is determined by microenvironment and origin. Brain and retinal microglia are both derived from yolk sac progenitors, yet their microenvironments differ. Utilizing single-cell RNA sequencing (scRNA-seq) data from mice, we tested the hypothesis that retinal and brain microglia exhibit distinct transcriptional profiles due to their unique microenvironments. Methods: Eyes and brains from 2-4 month wildtype mice were combined (20 eyes; 3 brains) to yield one biologically diverse sample per organ. Each tissue was digested into single cell suspensions, enriched for immune cells, and sorted for scRNA-seq. Analysis was performed in Seurat v3 including clustering, integration, and differential expression. Multi-parameter flow cytometry was used for validation of scRNA-seq results. Lymphocytic choriomeningitis virus (LCMV) Clone 13, which produces a systemic, chronic, and neurotropic infection, was used to validate scRNA-seq and flow cytometry results in vivo. Results: Cluster analysis of integrated gene expression data from eye and brain identified 6 Tmem119 + P2ry12 + microglial clusters. Differential expression analysis revealed that eye microglia were enriched for more pro-inflammatory processes including antigen processing via MHC class I (14.0-fold, H2-D1 and H2-K1) and positive regulation of T-cell immunity (8.4-fold) compared to brain microglia. Multi-parameter flow cytometry confirmed that retinal microglia expressed 3.2-fold greater H2-Db and 263.3-fold more H2-Kb than brain microglia. On Day 13 and 29 after LCMV infection, CD8+ T-cell density was greater in the retina than the brain. Discussion: Our data demonstrate that the microenvironment of retina and brain differs, resulting in microglia-specific gene expression changes. Specifically, retinal microglia express greater MHC class I by scRNA-seq and multi-parameter flow cytometry, resulting in a possibly enhanced capability to stimulate CD8+ T-cell inflammation during LCMV infection. These results may explain tissue-specific differences between retina and brain during systemic viral infections and CD8+ T-cell driven autoimmune disease.


Subject(s)
Brain , Microglia , Retina , Animals , Microglia/immunology , Microglia/metabolism , Mice , Retina/immunology , Retina/pathology , Brain/immunology , Brain/pathology , Brain/metabolism , Mice, Inbred C57BL , Lymphocytic choriomeningitis virus/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , T-Lymphocytes/immunology , Inflammation/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Single-Cell Analysis , CD8-Positive T-Lymphocytes/immunology , Transcriptome
12.
Bioengineering (Basel) ; 11(2)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38391606

ABSTRACT

In the modern era, patients often resort to the internet for answers to their health-related concerns, and clinics face challenges to providing timely response to patient concerns. This has led to a need to investigate the capabilities of AI chatbots for ophthalmic diagnosis and triage. In this in silico study, 80 simulated patient complaints in ophthalmology with varying urgency levels and clinical descriptors were entered into both ChatGPT and Bard in a systematic 3-step submission process asking chatbots to triage, diagnose, and evaluate urgency. Three ophthalmologists graded chatbot responses. Chatbots were significantly better at ophthalmic triage than diagnosis (90.0% appropriate triage vs. 48.8% correct leading diagnosis; p < 0.001), and GPT-4 performed better than Bard for appropriate triage recommendations (96.3% vs. 83.8%; p = 0.008), grader satisfaction for patient use (81.3% vs. 55.0%; p < 0.001), and lower potential harm rates (6.3% vs. 20.0%; p = 0.010). More descriptors improved the accuracy of diagnosis for both GPT-4 and Bard. These results indicate that chatbots may not need to recognize the correct diagnosis to provide appropriate ophthalmic triage, and there is a potential utility of these tools in aiding patients or triage staff; however, they are not a replacement for professional ophthalmic evaluation or advice.

13.
Am J Physiol Endocrinol Metab ; 305(5): E600-10, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23860123

ABSTRACT

Recently, a novel type 1 diabetes association locus was identified at human chromosome 6p31.3, and transcription factor 19 (TCF19) is a likely causal gene. Little is known about Tcf19, and we now show that it plays a role in both proliferation and apoptosis in insulinoma cells. Tcf19 is expressed in mouse and human islets, with increasing mRNA expression in nondiabetic obesity. The expression of Tcf19 is correlated with ß-cell mass expansion, suggesting that it may be a transcriptional regulator of ß-cell mass. Increasing proliferation and decreasing apoptotic cell death are two strategies to increase pancreatic ß-cell mass and prevent or delay diabetes. siRNA-mediated knockdown of Tcf19 in the INS-1 insulinoma cell line, a ß-cell model, results in a decrease in proliferation and an increase in apoptosis. There was a significant reduction in the expression of numerous cell cycle genes from the late G1 phase through the M phase, and cells were arrested at the G1/S checkpoint. We also observed increased apoptosis and susceptibility to endoplasmic reticulum (ER) stress after Tcf19 knockdown. There was a reduction in expression of genes important for the maintenance of ER homeostasis (Bip, p58(IPK), Edem1, and calreticulin) and an increase in proapoptotic genes (Bim, Bid, Nix, Gadd34, and Pdia2). Therefore, Tcf19 is necessary for both proliferation and survival and is a novel regulator of these pathways.


Subject(s)
Cell Cycle/physiology , Diabetes Mellitus/metabolism , Endoplasmic Reticulum Stress/physiology , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/physiology , Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Humans , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , RNA/chemistry , RNA/genetics , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription Factors/genetics
14.
JCI Insight ; 8(7)2023 04 10.
Article in English | MEDLINE | ID: mdl-36821388

ABSTRACT

Patients with neovascular AMD (nAMD) suffer vision loss from destructive angiogenesis, termed choroidal neovascularization (CNV). Macrophages are found in CNV lesions from patients with nAMD. Additionally, Ccr2-/- mice, which lack classical monocyte-derived macrophages, show reduced CNV size. However, macrophages are highly diverse cells that can perform multiple functions. We performed single-cell RNA-Seq on immune cells from WT and Ccr2-/- eyes to uncover macrophage heterogeneity during the laser-induced CNV mouse model of nAMD. We identified 12 macrophage clusters, including Spp1+ macrophages. Spp1+ macrophages were enriched from WT lasered eyes and expressed a proangiogenic transcriptome via multiple pathways, including vascular endothelial growth factor signaling, endothelial cell sprouting, cytokine signaling, and fibrosis. Additionally, Spp1+ macrophages expressed the marker CD11c, and CD11c+ macrophages were increased by laser and present in CNV lesions. Finally, CD11c+ macrophage depletion reduced CNV size by 40%. These findings broaden our understanding of ocular macrophage heterogeneity and implicate CD11c+ macrophages as potential therapeutic targets for treatment-resistant patients with nAMD.


Subject(s)
Choroidal Neovascularization , Wet Macular Degeneration , Animals , Mice , Angiogenesis Inhibitors/therapeutic use , Choroidal Neovascularization/drug therapy , Macrophages/metabolism , Vascular Endothelial Growth Factor A/metabolism , Visual Acuity , Wet Macular Degeneration/pathology , CD11c Antigen/metabolism
15.
JCI Insight ; 8(23)2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37917183

ABSTRACT

The management of preretinal fibrovascular membranes, a devastating complication of advanced diabetic retinopathy (DR), remains challenging. We characterized the molecular profile of cell populations in these fibrovascular membranes to identify potentially new therapeutic targets. Preretinal fibrovascular membranes were surgically removed from patients and submitted for single-cell RNA-Seq (scRNA-Seq). Differential gene expression was implemented to define the transcriptomics profile of these cells and revealed the presence of endothelial, inflammatory, and stromal cells. Endothelial cell reclustering identified subclusters characterized by noncanonical transcriptomics profile and active angiogenesis. Deeper investigation of the inflammatory cells showed a subcluster of macrophages expressing proangiogenic cytokines, presumably contributing to angiogenesis. The stromal cell cluster included a pericyte-myofibroblast transdifferentiating subcluster, indicating the involvement of pericytes in fibrogenesis. Differentially expressed gene analysis showed that Adipocyte Enhancer-binding Protein 1, AEBP1, was significantly upregulated in myofibroblast clusters, suggesting that this molecule may have a role in transformation. Cell culture experiments with human retinal pericytes (HRP) in high-glucose condition confirmed the molecular transformation of pericytes toward myofibroblastic lineage. AEBP1 siRNA transfection in HRP reduced the expression of profibrotic markers in high glucose. In conclusion, AEBP1 signaling modulates pericyte-myofibroblast transformation, suggesting that targeting AEBP1 could prevent scar tissue formation in advanced DR.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Humans , Diabetic Retinopathy/metabolism , Retina/metabolism , Pericytes/metabolism , Glucose/metabolism , Gene Expression Profiling , Diabetes Mellitus/metabolism , Carboxypeptidases/metabolism , Repressor Proteins/genetics
16.
Ophthalmol Retina ; 7(9): 745-752, 2023 09.
Article in English | MEDLINE | ID: mdl-37146684

ABSTRACT

OBJECTIVE: To determine whether levodopa (L-DOPA) is associated with a reduced likelihood of developing neovascular age-related macular degeneration (AMD). DESIGN: Three studies were performed: retrospective analyses in the Vestrum Health Retina Database (#1-2) and case-control analysis in the Merative MarketScan Research Databases (#3). PARTICIPANTS: Eyes with neovascular AMD and 2 years of follow-up (#1). Eyes with non-neovascular AMD and 1 to 5 years of follow-up (#2). Patients aged ≥ 55 years with newly diagnosed neovascular AMD matched to controls without neovascular AMD (#3). METHODS: Eyes were divided into 2 groups (#1-2): exposed to L-DOPA before or on the date of neovascular (#1) or nonneovascular (#2) AMD diagnosis, and eyes not exposed to L-DOPA. We extracted AMD risk factors, number of intravitreal injections (#1), and conversion rate to neovascular AMD (#2). We calculated the percentage of newly diagnosed neovascular AMD cases and matched controls exposed to any L-DOPA and determined the cumulative 2-year dose in grams by tertiles (< 100 mg, approximately 100-300 mg, and approximately > 300 mg per day, #3). MAIN OUTCOME MEASURES: Number of intravitreal injections (#1) and detection of new-onset neovascular AMD (#2-3) after adjusting for AMD risk factors. RESULTS: In the Vestrum database, eyes with neovascular AMD that were exposed to L-DOPA underwent 1 fewer intravitreal injection over 2 years (N = 84 088 control vs. 530 L-DOPA eyes, P = 0.006). In eyes with nonneovascular AMD (N = 42 081-203 155 control vs. 314-1525 L-DOPA eyes), L-DOPA exposure was associated with a reduced risk of conversion to neovascular AMD by 21% at year 2 (P = 0.029), 35% at years 3 to 4 (P < 0.001), and 28% at year 5 (P = 0.024). In the MarketScan databases (N = 86 900 per group), cumulative 2-year doses of L-DOPA between approximately 100 to 300 mg per day and approximately > 300 mg were associated with decreased odds of developing neovascular AMD by 15% (odds ratio [OR], 0.85; 95% confidence interval [CI], 0.75-0.97) and 23% (OR, 0.77; 95% CI, 0.67-0.87), respectively. CONCLUSIONS: Levodopa use was associated with reduced detection of new-onset neovascular AMD. A prospective, randomized clinical trial should be considered to investigate whether low-dose L-DOPA reduces neovascular AMD conversion. FINANCIAL DISCLOSURE(S): Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.


Subject(s)
Levodopa , Macular Degeneration , Humans , Levodopa/therapeutic use , Retrospective Studies , Prospective Studies , Eye
17.
Curr Protoc ; 3(4): e739, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37078364

ABSTRACT

Inflammation is the body's response to injury and harmful stimuli and contributes to a range of infectious and noninfectious diseases. Inflammation occurs through a series of well-defined leukocyte-endothelial cell interactions, including rolling, activation, adhesion, transmigration, and subsequent migration through the extracellular matrix. Being able to visualize the stages of inflammation is important for a better understanding of its role in diseases processes. Detailed in this article are protocols for imaging immune cell infiltration and transendothelial migration in vascular tissue beds, including those in the mouse ear, cremaster muscle, brain, lung, and retina. Also described are protocols for inducing inflammation and quantifying leukocytes with FIJI imaging software. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Induction of croton oil dermatitis Alternate Protocol 1: Induction of croton oil dermatitis using genetically fluorescent mice Basic Protocol 2: Intravital microscopy of the mouse cremaster muscle Support Protocol: Making a silicone stage Basic Protocol 3: Wide-field microscopy of the mouse brain Basic Protocol 4: Imaging the lungs (ex vivo) Alternate Protocol 2: Inflating the lungs without tracheostomy Basic Protocol 5: Inducing, imaging, and quantifying infiltration of leukocytes in mouse retina.


Subject(s)
Dermatitis , Transendothelial and Transepithelial Migration , Mice , Animals , Croton Oil , Leukocytes/physiology , Inflammation/diagnostic imaging
18.
Sci Adv ; 9(41): eadi8534, 2023 10 13.
Article in English | MEDLINE | ID: mdl-37831763

ABSTRACT

Neovascular age-related macular degeneration (nAMD) is the leading cause of blindness in the developed world. Current therapy includes monthly intraocular injections of anti-VEGF antibodies, which are ineffective in up to one third of patients. Thrombospondin-1 (TSP1) inhibits angiogenesis via CD36 binding, and its down-regulated expression is negatively associated with the onset of nAMD. Here, we describe TSP1 mimetic protein-like polymers (TSP1 PLPs). TSP1 PLPs bind CD36 with high affinity, resist degradation, show prolonged intraocular half-lives (13.1 hours), have no toxicity at relevant concentrations in vivo (40 µM), and are more efficacious in ex vivo choroidal sprouting assays compared to the peptide sequence and Eylea (aflibercept), the current standard of care anti-VEGF treatment. Furthermore, PLPs exhibit superior in vivo efficacy in a mouse model for nAMD compared to control PLPs consisting of scrambled peptide sequences, using fluorescein angiography and immunofluorescence. Since TSP-1 inhibits angiogenesis by VEGF-dependent and independent mechanisms, TSP1 PLPs are a potential therapeutic for patients with anti-VEGF treatment-resistant nAMD.


Subject(s)
Macular Degeneration , Ranibizumab , Animals , Mice , Humans , Ranibizumab/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Thrombospondin 1/therapeutic use , Macular Degeneration/drug therapy , Peptides
20.
Sci Rep ; 12(1): 12864, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35896600

ABSTRACT

The identity of vitreoretinal interface macrophage-like cells (MLCs) remains unknown and potential candidates include retinal microglia, perivascular macrophages, monocyte-derived macrophages, and/or vitreal hyalocytes. Since hyalocytes are detectable on the posterior vitreous surface after vitreous extraction in animals, we imaged patients with and without posterior vitreous detachment (PVD) to determine if hyalocytes are the principal MLC component. We performed repeated foveal-centered 3 × 3 mm OCT-A images from 21 eyes (11 no PVD and 10 PVD eyes). Images were registered, segmented, and averaged. The OCT slab from 0 to 3 microns above the internal limiting membrane was used to detect MLCs. We calculated MLC density and distribution in relation to the superficial vascular plexus for 3 vascular regions-on vessels, perivascular, and non-vascular. MLC density was 1.8-fold greater in the PVD group compared to the no PVD group (P = 0.04). MLCs in eyes with PVD were increased 1.9-fold on-vessel (P = 0.07), 1.9-fold in the perivascular region (P = 0.12), and 2.2-fold in non-vascular areas (P = 0.22). MLC density was not severely reduced after PVD, suggesting that the majority of MLCs are not vitreal hyalocytes. PVD status is an important parameter in future MLC studies.


Subject(s)
Retinal Detachment , Vitreous Detachment , Animals , Macrophages , Retina
SELECTION OF CITATIONS
SEARCH DETAIL