Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Neuroinflammation ; 20(1): 238, 2023 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-37858232

RESUMEN

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.


Asunto(s)
Neovascularización Coroidal , Degeneración Macular , Animales , Ratones , Neovascularización Coroidal/genética , Modelos Animales de Enfermedad , Macrófagos/fisiología , Degeneración Macular/genética , Ratones Endogámicos C57BL , Microglía , Monocitos
2.
Dis Model Mech ; 16(7)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37401371

RESUMEN

Oxidative stress has been implicated in the pathogenesis of age-related macular degeneration, the leading cause of blindness in older adults, with retinal pigment epithelium (RPE) cells playing a key role. To better understand the cytotoxic mechanisms underlying oxidative stress, we used cell culture and mouse models of iron overload, as iron can catalyze reactive oxygen species formation in the RPE. Iron-loading of cultured induced pluripotent stem cell-derived RPE cells increased lysosomal abundance, impaired proteolysis and reduced the activity of a subset of lysosomal enzymes, including lysosomal acid lipase (LIPA) and acid sphingomyelinase (SMPD1). In a liver-specific Hepc (Hamp) knockout murine model of systemic iron overload, RPE cells accumulated lipid peroxidation adducts and lysosomes, developed progressive hypertrophy and underwent cell death. Proteomic and lipidomic analyses revealed accumulation of lysosomal proteins, ceramide biosynthetic enzymes and ceramides. The proteolytic enzyme cathepsin D (CTSD) had impaired maturation. A large proportion of lysosomes were galectin-3 (Lgals3) positive, suggesting cytotoxic lysosomal membrane permeabilization. Collectively, these results demonstrate that iron overload induces lysosomal accumulation and impairs lysosomal function, likely due to iron-induced lipid peroxides that can inhibit lysosomal enzymes.


Asunto(s)
Sobrecarga de Hierro , Proteómica , Ratones , Animales , Estrés Oxidativo , Lisosomas/metabolismo , Hierro/metabolismo , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Células Epiteliales/metabolismo , Pigmentos Retinianos/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
3.
Front Cell Neurosci ; 17: 1156829, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37362000

RESUMEN

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.
Br J Ophthalmol ; 107(2): 215-220, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34413054

RESUMEN

BACKGROUND/AIMS: Glucagon-like peptide-1 receptor (GLP-1R) agonists regulate blood glucose and are commonly used to treat type 2 diabetes mellitus. Recent work showed that treatment with the GLP-1R agonist NLY01 decreased retinal neuroinflammation and glial activation to rescue retinal ganglion cells in a mouse model of glaucoma. In this study, we used an insurance claims database (Clinformatics Data Mart) to examine whether GLP-1R agonist exposure impacts glaucoma risk. METHODS: A retrospective cohort of patients who initiated a new GLP-1R agonist was 1:3 age, gender, race, classes of active diabetes medications and year of index date matched to patients who initiated a different class of oral diabetic medication. Inverse probability of treatment weighting (IPTW) was used within a multivariable Cox proportional hazard regression model to test the association between GLP-1R agonist exposure and a new diagnosis of primary open-angle glaucoma, glaucoma suspect or low-tension glaucoma. RESULTS: Cohorts were comprised of 1961 new users of GLP-1R agonists matched to 4371 unexposed controls. After IPTW, all variables were balanced (standard mean deviation <|0.1|) between cohorts. Ten (0.51%) new diagnoses of glaucoma were present in the GLP-1R agonist cohort compared with 58 (1.33%) in the unexposed controls. After adjustment, GLP-1R exposure conferred a reduced hazard of 0.56 (95% CI: 0.36 to 0.89, p=0.01), suggesting that GLP-1R agonists decrease the risk for glaucoma. CONCLUSIONS: GLP-1R agonist use was associated with a statistically significant hazard reduction for a new diagnosis of glaucoma. Our findings support further investigations into the use of GLP-1R agonists in glaucoma prevention.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glaucoma de Ángulo Abierto , Animales , Ratones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Péptido 1 Similar al Glucagón/uso terapéutico , Estudios Retrospectivos , Glaucoma de Ángulo Abierto/tratamiento farmacológico , Glaucoma de Ángulo Abierto/prevención & control , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/uso terapéutico
5.
Cell Rep ; 39(11): 110942, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705048

RESUMEN

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.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Tejido Adiposo/metabolismo , Humanos , Hierro/metabolismo , Degeneración Macular/metabolismo , Estrés Oxidativo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
6.
Exp Eye Res ; 218: 109028, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35271829

RESUMEN

Iron accumulation has been implicated in degenerative retinal diseases. It can catalyze the production of damaging reactive oxygen species. Previous work has demonstrated iron accumulation in multiple retinal diseases, including age-related macular degeneration and diabetic retinopathy. In mice, systemic knockout of the ferroxidases ceruloplasmin (Cp) and hephaestin (Heph), which oxidize iron, results in retinal iron accumulation and iron-induced degeneration. To determine the role of Heph in the retina, we generated a neural retina-specific Heph knockout on a background of systemic Cp knockout. This resulted in elevated neural retina iron. Conversely, retinal ganglion cells had elevated transferrin receptor and decreased ferritin, suggesting diminished iron levels. The retinal degeneration observed in systemic Cp-/-, Heph-/- mice did not occur. These findings indicate that Heph has a local role in regulating neural retina iron homeostasis, but also suggest that preserved Heph function in either the RPE or systemically mitigates the degeneration phenotype observed in the systemic Cp-/-, Heph-/- mice.


Asunto(s)
Degeneración Macular , Proteínas de la Membrana , Animales , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Homeostasis , Hierro/metabolismo , Degeneración Macular/genética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Retina/metabolismo
7.
Exp Eye Res ; 218: 108988, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35202704

RESUMEN

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.


Asunto(s)
Proteínas de Transporte de Catión , Sobrecarga de Hierro , Animales , Proteínas de Transporte de Catión/genética , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Hierro/metabolismo , Ratones , Ratones Noqueados , Retina/metabolismo
8.
Cell Rep ; 38(7): 110358, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35172141

RESUMEN

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.


Asunto(s)
Interleucina-6/metabolismo , Hierro/metabolismo , Neuronas/metabolismo , alfa-Sinucleína/toxicidad , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/patología , Femenino , Quelantes del Hierro/farmacología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Transducción de Señal/efectos de los fármacos , Sustancia Negra/efectos de los fármacos , Sustancia Negra/patología
9.
Aging Cell ; 20(11): e13490, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34626070

RESUMEN

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.


Asunto(s)
Compuestos Férricos/administración & dosificación , Atrofia Geográfica/inducido químicamente , Atrofia Geográfica/complicaciones , Inyecciones Intraoculares/métodos , Oftalmía Simpática/inducido químicamente , Oftalmía Simpática/complicaciones , Estrés Oxidativo/efectos de los fármacos , Compuestos de Amonio Cuaternario/administración & dosificación , Animales , Modelos Animales de Enfermedad , Atrofia Geográfica/diagnóstico por imagen , Atrofia Geográfica/metabolismo , Hierro/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oftalmía Simpática/diagnóstico por imagen , Oftalmía Simpática/metabolismo , Imagen Óptica/métodos , Epitelio Pigmentado de la Retina/diagnóstico por imagen , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología
10.
Cell Rep ; 33(5): 108271, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33147455

RESUMEN

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.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Inflamación/patología , Hipertensión Ocular/complicaciones , Neuronas Retinianas/patología , Animales , Astrocitos/patología , Antígeno CD11b/metabolismo , Muerte Celular , Complemento C1q/metabolismo , Femenino , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Interleucina-1alfa/metabolismo , Presión Intraocular , Masculino , Ratones Endogámicos C57BL , Hipertensión Ocular/fisiopatología , Células Ganglionares de la Retina/patología , Factor de Necrosis Tumoral alfa/metabolismo
11.
Am J Pathol ; 189(9): 1814-1830, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31287995

RESUMEN

The liver secretes hepcidin (Hepc) into the bloodstream to reduce blood iron levels. Hepc accomplishes this by triggering degradation of the only known cellular iron exporter ferroportin in the gut, macrophages, and liver. We previously demonstrated that systemic Hepc knockout (HepcKO) mice, which have high serum iron, develop retinal iron overload and degeneration. However, it was unclear whether this is caused by high blood iron levels or, alternatively, retinal iron influx that would normally be regulated by retina-produced Hepc. To address this question, retinas of liver-specific and retina-specific HepcKO mice were studied. Liver-specific HepcKO mice had elevated blood and retinal pigment epithelium (RPE) iron levels and increased free (labile) iron levels in the retina, despite an intact blood-retinal barrier. This led to RPE hypertrophy associated with lipofuscin-laden lysosome accumulation. Photoreceptors also degenerated focally. In contrast, there was no change in retinal or RPE iron levels or degeneration in the retina-specific HepcKO mice. These data indicate that high blood iron levels can lead to retinal iron accumulation and degeneration. High blood iron levels can occur in patients with hereditary hemochromatosis or result from use of iron supplements or multiple blood transfusions. Our results suggest that high blood iron levels may cause or exacerbate retinal disease.


Asunto(s)
Hepcidinas/fisiología , Sobrecarga de Hierro/etiología , Hierro/metabolismo , Hígado/metabolismo , Retina/metabolismo , Degeneración Retiniana/etiología , Animales , Barrera Hematorretinal , Femenino , Sobrecarga de Hierro/metabolismo , Sobrecarga de Hierro/patología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Retina/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología
12.
Invest Ophthalmol Vis Sci ; 58(10): 4223-4234, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28846772

RESUMEN

Purpose: Retinal iron accumulation is observed in a wide range of retinal degenerative diseases, including AMD. Previous work suggests that Müller glial cells may be important mediators of retinal iron transport, distribution, and regulation. A transgenic model of Müller cell loss recently demonstrated that primary Müller cell ablation leads to blood-retinal barrier leakage and photoreceptor degeneration, and it recapitulates clinical features observed in macular telangiectasia type 2 (MacTel2), a rare human disease that features Müller cell loss. We used this mouse model to determine the effect of Müller cell loss on retinal iron homeostasis. Methods: Changes in total retinal iron levels after Müller cell ablation were measured using inductively coupled plasma mass spectrometry. Corresponding changes in the expression of iron flux and iron storage proteins were determined using quantitative PCR, Western analysis, and immunohistochemistry. Results: Müller cell loss led to blood-retinal barrier breakdown and increased iron levels throughout the neurosensory retina. There were corresponding changes in mRNA and/or protein levels of ferritin, transferrin receptor, ferroportin, Zip8, and Zip14. There were also increased iron levels within the RPE of retinal sections from a patient with MacTel2 and both RPE and neurosensory retina of a patient with diabetic retinopathy, which, like MacTel2, causes retinal vascular leakage. Conclusion: This study shows that Müller cells and the blood-retinal barrier play pivotal roles in the regulation of retinal iron homeostasis. The retinal iron accumulation resulting from blood-retinal barrier dysfunction may contribute to retinal degeneration in this model and in diseases such as MacTel2 and diabetic retinopathy.


Asunto(s)
Modelos Animales de Enfermedad , Células Ependimogliales/patología , Hierro/metabolismo , Retina/metabolismo , Telangiectasia Retiniana/metabolismo , Anciano , Animales , Barrera Hematorretinal/metabolismo , Barrera Hematorretinal/patología , Western Blotting , Permeabilidad Capilar , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Femenino , Ferritinas/genética , Ferritinas/metabolismo , Humanos , Inmunohistoquímica , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Persona de Mediana Edad , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Telangiectasia Retiniana/genética
13.
Mol Vis ; 23: 210-218, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28442885

RESUMEN

PURPOSE: The complement system is involved in the pathogenesis of age-related macular degeneration (AMD). Because activated microglia are also associated with AMD, we studied the relationship between complement anaphylatoxin receptors and microglial recruitment. METHODS: We assessed the effect of anaphylatoxin C3a receptor (C3aR) and C5a receptor (C5aR) knockout (KO) on light damage-induced migration of microglia/macrophages into the mouse outer retina via immunofluorescence and real-time quantitative PCR. RESULTS: We found that the mRNA levels of C3, C5, C3aR, C5aR, and two activators of the complement alternative pathway, Cfb and Cfd, were all upregulated after light exposure. Retinal Iba1-positive microglia/macrophages express receptors for C3a and C5a. Light damage increased the number of retinal Iba1-positive cells and the mRNA levels of Iba1. Compared with the wild-type (WT) mice, these increases were attenuated in the C5aR KO mice but not in the C3aR KO mice. CONCLUSIONS: C5aR but not C3aR promoted the recruitment of microglia/macrophages. These divergent properties of complement anaphylatoxins in the light damage model provide a rationale for testing the differential effects of these receptors in additional retinal and neurodegeneration models.


Asunto(s)
Movimiento Celular/efectos de la radiación , Técnicas de Inactivación de Genes , Luz/efectos adversos , Macrófagos/fisiología , Microglía/fisiología , Receptor de Anafilatoxina C5a/genética , Degeneración Retiniana/patología , Animales , Proteínas de Unión al Calcio/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas de Microfilamentos/metabolismo , ARN Mensajero/genética , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/genética , Retina/efectos de la radiación , Degeneración Retiniana/etiología
14.
Exp Eye Res ; 155: 15-23, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28057442

RESUMEN

Intracellular retinal iron accumulation has been implicated in the pathogenesis of age-related macular degeneration (AMD), the leading cause of irreversible blindness among individuals over the age of 50. Ceruloplasmin/hephaestin double knockout mice (Cp/Heph DKO) and hepcidin knockout mice (Hepc KO) accumulate retinal iron and model some features of AMD. Two canonical pathways govern cellular iron import - transferrin-bound iron import and non-transferrin bound iron import. In Cp/Heph DKO and Hepc KO iron-loaded retinas, transferrin-bound iron import is downregulated. Despite this effort to reduce cellular iron burden, iron continues to accumulate in these retinas in an age-dependent manner. Quantitative RT-PCR and Western analysis were used to quantify the expression of three ferrous iron importers, Dmt1, Zip8, and Zip14, in wild-type (Wt), Cp/Heph DKO, and Hepc KO retinas. Zip8 and Zip14 protein levels were analyzed using Western analysis in mice injected intravitreally with either apo- or holo-transferrin to elucidate one possible mechanism of Zip14 regulation in the retina. Both zip8 and zip14 were expressed in the mouse retina. Paradoxically, protein levels of non-transferrin bound iron importers were upregulated in both Cp/Heph DKO and Hepc KO retinas. Intravitreal holo-transferrin injection decreased Zip 14 protein levels. These data indicate that Zip8 and Zip14 may take up increasing amounts of non-transferrin bound iron in these two mouse models of retinal iron accumulation. Their upregulation in these already iron-loaded retinas suggests a vicious cycle leading to toxicity.


Asunto(s)
Proteínas de Transporte de Catión/genética , Regulación de la Expresión Génica , Hierro/metabolismo , Degeneración Macular/metabolismo , ARN/genética , Retina/metabolismo , Animales , Western Blotting , Proteínas de Transporte de Catión/biosíntesis , Modelos Animales de Enfermedad , Inyecciones Intravítreas , Degeneración Macular/genética , Degeneración Macular/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/efectos de los fármacos , Retina/patología , Transferrina/administración & dosificación
15.
FASEB J ; 30(2): 813-23, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26506980

RESUMEN

Because ferroportin (Fpn) is the only known mammalian cellular iron exporter, understanding its localization and regulation within the retina would shed light on the direction of retinal iron flux. The hormone hepcidin may regulate retinal Fpn, as it triggers Fpn degradation in the gut. Immunofluorescence was used to label Fpn in retinas of mice with 4 different genotypes (wild type; Fpn C326S, a hepcidin-resistant Fpn; hepcidin knockout; and ceruloplasmin/hephaestin double knockout). No significant difference in Fpn levels was observed in these retinas. Fpn localized to the abluminal side of the outer plexiform vascular endothelial cells, Müller glia cells, and the basolateral side of the retinal pigment epithelium. Adeno-associated virus (AAV)-hepcidin was injected into the eyes of hepcidin knockout mice, while AAV-lacZ was injected into the contralateral eyes as a control. AAV-hepcidin injected eyes had increased ferritin immunolabeling in retinal vascular endothelial cells. Fpn C326S mice had systemic iron overload compared to wild type and had the fastest retinal iron accumulation of any hereditary model studied to date. The results suggest that physiologic hepcidin levels are insufficient to alter Fpn levels within the retinal pigment epithelium and Müller cells, but may limit iron transport into the retina from vascular endothelial cells.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Hepcidinas/metabolismo , Hierro/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Sustitución de Aminoácidos , Animales , Proteínas de Transporte de Catión/genética , Hepcidinas/genética , Ratones , Ratones Noqueados , Mutación Missense , Epitelio Pigmentado de la Retina/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...