Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
J Clin Invest ; 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39207852

ABSTRACT

The blood-retina barrier (BRB), which is disrupted in diabetic retinopathy (DR) and uveitis, is an important anatomical characteristic of the retina, regulating nutrient, waste, water, protein, and immune cell flux. The BRB is composed of endothelial cell tight junctions, pericytes, astrocyte end feet, a collagen basement membrane, and perivascular macrophages. Despite the importance of the BRB, retinal perivascular macrophage function remains unknown. We found that retinal perivascular macrophages reside on post-capillary venules in the superficial vascular plexus and express MHCII. Using single-cell RNA-sequencing, we found that perivascular macrophages express a pro-chemotactic transcriptome and identified Pf4/CXCL4 as a perivascular macrophage marker. We used Pf4Cre mice to specifically deplete perivascular macrophages. To model retinal inflammation, we performed intraocular CCL2 injections. Ly6C+ monocytes crossed the BRB proximal to perivascular macrophages. Depletion of perivascular macrophages severely hampered Ly6C+ monocyte infiltration. These data suggest that retinal perivascular macrophages orchestrate immune cell migration across the BRB, with implications for inflammatory ocular diseases including DR and uveitis.

2.
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
3.
Front Cell Neurosci ; 17: 1156829, 2023.
Article in English | MEDLINE | ID: mdl-37362000

ABSTRACT

Glaucomatous neurodegeneration, a blinding disease affecting millions worldwide, has a need for the exploration of new and effective therapies. Previously, the glucagon-like peptide-1 receptor (GLP-1R) agonist NLY01 was shown to reduce microglia/macrophage activation, rescuing retinal ganglion cells after IOP elevation in an animal model of glaucoma. GLP-1R agonist use is also associated with a reduced risk for glaucoma in patients with diabetes. In this study, we demonstrate that several commercially available GLP-1R agonists, administered either systemically or topically, hold protective potential in a mouse model of hypertensive glaucoma. Further, the resulting neuroprotection likely occurs through the same pathways previously shown for NLY01. This work contributes to a growing body of evidence suggesting that GLP-1R agonists represent a viable therapeutic option for glaucoma.

4.
Cell Rep ; 39(11): 110942, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35705048

ABSTRACT

Age-related macular degeneration (AMD), the leading cause of irreversible blindness among Americans over 50, is characterized by dysfunction and death of retinal pigment epithelial (RPE) cells. The RPE accumulates iron in AMD, and iron overload triggers RPE cell death in vitro and in vivo. However, the mechanism of RPE iron accumulation in AMD is unknown. We show that high-fat-diet-induced obesity, a risk factor for AMD, drives systemic and local inflammatory circuits upregulating interleukin-1ß (IL-1ß). IL-1ß upregulates RPE iron importers and downregulates iron exporters, causing iron accumulation, oxidative stress, and dysfunction. We term this maladaptive, chronic activation of a nutritional immunity pathway the cellular iron sequestration response (CISR). RNA sequencing (RNA-seq) analysis of choroid and retina from human donors revealed that hallmarks of this pathway are present in AMD microglia and macrophages. Together, these data suggest that inflamed adipose tissue, through the CISR, can lead to RPE pathology in AMD.


Subject(s)
Macular Degeneration , Retinal Pigment Epithelium , Adipose Tissue/metabolism , Humans , Iron/metabolism , Macular Degeneration/metabolism , Oxidative Stress , Retina/metabolism , Retinal Pigment Epithelium/metabolism
5.
Cell Rep ; 38(7): 110358, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172141

ABSTRACT

α-synuclein (α-syn) aggregation and accumulation drive neurodegeneration in Parkinson's disease (PD). The substantia nigra of patients with PD contains excess iron, yet the underlying mechanism accounting for this iron accumulation is unclear. Here, we show that misfolded α-syn activates microglia, which release interleukin 6 (IL-6). IL-6, via its trans-signaling pathway, induces changes in the neuronal iron transcriptome that promote ferrous iron uptake and decrease cellular iron export via a pathway we term the cellular iron sequestration response, or CISR. The brains of patients with PD exhibit molecular signatures of the IL-6-mediated CISR. Genetic deletion of IL-6, or treatment with the iron chelator deferiprone, reduces pathological α-syn toxicity in a mouse model of sporadic PD. These data suggest that IL-6-induced CISR leads to toxic neuronal iron accumulation, contributing to synuclein-induced neurodegeneration.


Subject(s)
Interleukin-6/metabolism , Iron/metabolism , Neurons/metabolism , alpha-Synuclein/toxicity , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Female , Iron Chelating Agents/pharmacology , Male , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/pathology , Parkinson Disease/genetics , Parkinson Disease/pathology , Signal Transduction/drug effects , Substantia Nigra/drug effects , Substantia Nigra/pathology
6.
Exp Eye Res ; 218: 108988, 2022 05.
Article in English | MEDLINE | ID: mdl-35202704

ABSTRACT

Iron-induced oxidative stress can cause or exacerbate retinal degenerative diseases. Retinal iron overload has been reported in several mouse disease models with systemic or neural retina-specific knockout (KO) of homologous ferroxidases ceruloplasmin (Cp) and hephaestin (Heph). Cp and Heph can potentiate ferroportin (Fpn) mediated cellular iron export. Here, we used retina-specific Fpn KO mice to test the hypothesis that retinal iron overload in Cp/Heph DKO mice is caused by impaired iron export from neurons and glia. Surprisingly, there was no indication of retinal iron overload in retina-specific Fpn KO mice: the mRNA levels of transferrin receptor in the retina were not altered at 7-10-months age. Consistent with this, levels and localization of ferritin light chain were unchanged. To "stress the system", we injected iron intraperitoneally into Fpn KO mice with or without Cp KO. Only mice with both retina-specific Fpn KO and Cp KO had modestly elevated retinal iron levels. These results suggest that impaired iron export through Fpn is not sufficient to explain the retinal iron overload in Cp/Heph DKO mice. An increase in the levels of retinal ferrous iron caused by the absence of these ferroxidases, followed by uptake into cells by ferrous iron importers, is most likely necessary.


Subject(s)
Cation Transport Proteins , Iron Overload , Animals , Cation Transport Proteins/genetics , Ceruloplasmin/genetics , Ceruloplasmin/metabolism , Iron/metabolism , Mice , Mice, Knockout , Retina/metabolism
7.
Aging Cell ; 20(11): e13490, 2021 11.
Article in English | MEDLINE | ID: mdl-34626070

ABSTRACT

Iron has been implicated in the pathogenesis of age-related retinal diseases, including age-related macular degeneration (AMD). Previous work showed that intravitreal (IVT) injection of iron induces acute photoreceptor death, lipid peroxidation, and autofluorescence (AF). Herein, we extend this work, finding surprising chronic features of the model: geographic atrophy and sympathetic ophthalmia. We provide new mechanistic insights derived from focal AF in the photoreceptors, quantification of bisretinoids, and localization of carboxyethyl pyrrole, an oxidized adduct of docosahexaenoic acid associated with AMD. In mice given IVT ferric ammonium citrate (FAC), RPE died in patches that slowly expanded at their borders, like human geographic atrophy. There was green AF in the photoreceptor ellipsoid, a mitochondria-rich region, 4 h after injection, followed later by gold AF in rod outer segments, RPE and subretinal myeloid cells. The green AF signature is consistent with flavin adenine dinucleotide, while measured increases in the bisretinoid all-trans-retinal dimer are consistent with the gold AF. FAC induced formation carboxyethyl pyrrole accumulation first in photoreceptors, then in RPE and myeloid cells. Quantitative PCR on neural retina and RPE indicated antioxidant upregulation and inflammation. Unexpectedly, reminiscent of sympathetic ophthalmia, autofluorescent myeloid cells containing abundant iron infiltrated the saline-injected fellow eyes only if the contralateral eye had received IVT FAC. These findings provide mechanistic insights into the potential toxicity caused by AMD-associated retinal iron accumulation. The mouse model will be useful for testing antioxidants, iron chelators, ferroptosis inhibitors, anti-inflammatory medications, and choroidal neovascularization inhibitors.


Subject(s)
Ferric Compounds/administration & dosage , Geographic Atrophy/chemically induced , Geographic Atrophy/complications , Injections, Intraocular/methods , Ophthalmia, Sympathetic/chemically induced , Ophthalmia, Sympathetic/complications , Oxidative Stress/drug effects , Quaternary Ammonium Compounds/administration & dosage , Animals , Disease Models, Animal , Geographic Atrophy/diagnostic imaging , Geographic Atrophy/metabolism , Iron/metabolism , Male , Mice , Mice, Inbred C57BL , Ophthalmia, Sympathetic/diagnostic imaging , Ophthalmia, Sympathetic/metabolism , Optical Imaging/methods , Retinal Pigment Epithelium/diagnostic imaging , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology
8.
Cell Rep ; 33(5): 108271, 2020 11 03.
Article in English | MEDLINE | ID: mdl-33147455

ABSTRACT

Glaucoma is the leading cause of irreversible blindness and is characterized by the death of retinal ganglion cells (RGCs). Recent studies have implicated pro-inflammatory microglia, macrophages, and A1 astrocytes in the pathogenesis of neurodegenerative diseases. The role of pro-inflammatory, neurotoxic A1 astrocytes in glaucoma is just beginning to be explored. Using a mouse model of glaucoma, we demonstrate that ocular hypertension is sufficient to trigger production of C1q, interleukin-1α (IL-1α), and tumor necrosis factor α (TNF-α), three cytokines necessary and sufficient to drive the formation of A1 astrocytes. Upregulation of these cytokines occurs first in CD11b+ CD11c+ cells followed by CD11b+ CD11c- cells. Ablation of this pathway, by either genetic deletions of C1qa, IL-1α, and TNF-α, or treatment with glucagon-like peptide-1 receptor agonist NLY01, reduces A1 astrocyte transformation and RGC death. Together, these results highlight a neuroinflammatory mechanism of glaucomatous neurodegeneration that can be therapeutically targeted by NLY01 administration.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Inflammation/pathology , Ocular Hypertension/complications , Retinal Neurons/pathology , Animals , Astrocytes/pathology , CD11b Antigen/metabolism , Cell Death , Complement C1q/metabolism , Female , Glucagon-Like Peptide-1 Receptor/metabolism , Interleukin-1alpha/metabolism , Intraocular Pressure , Male , Mice, Inbred C57BL , Ocular Hypertension/physiopathology , Retinal Ganglion Cells/pathology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL