Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Immunol ; 25(1): 31, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38734625

RESUMEN

BACKGROUND: Thyroid eye disease (TED) is an inflammatory process involving lymphocyte-mediated immune response and orbital tissue damage. The anti-insulin-like growth factor-1 receptor (IGF-1R) antibodies produced by B lymphocytes are involved in the activation of orbital fibroblasts and the inflammatory process of orbital tissue damage in TED. The purpose of this study was to explore the role of IGF-1R in the mechanistic connection between orbital fibroblasts and B lymphocytes in TED. METHODS: Orbital fibroblasts sampled from orbital connective tissues and peripheral B lymphocytes isolated from peripheral blood, which were obtained from 15 patients with TED and 15 control patients, were co-cultured at a ratio of 1:20. The level of IGF-1R expression in orbital fibroblasts was evaluated by flow cytometry and confocal microscopy. Transient B lymphocyte depletion was induced with anti-CD20 monoclonal antibody rituximab, while the IGF-1R pathway was blocked by the IGF-1R binding protein. The expression levels of interleukin-6 (IL-6) and regulated upon activation, normal T cell expressed and secreted (RANTES) in the co-culture model were quantified via ELISA. RESULTS: IGF-1R expression was significantly elevated in TED orbital fibroblasts compared to that of controls. A 24-h co-culture of orbital fibroblasts with peripheral B lymphocytes induced elevated expression levels of IL-6 and RANTES in each group (TED patients and controls), with the highest levels occurring in TED patients (T + T group). Rituximab and IGF-1R binding protein significantly inhibited increased levels of IL-6 and RANTES in the co-culture model of TED patients. CONCLUSIONS: IGF-1R may mediate interaction between orbital fibroblasts and peripheral B lymphocytes; thus, blocking IGF-1R may reduce the local inflammatory response in TED. Rituximab-mediated B lymphocyte depletion played a role in inhibiting inflammatory responses in this in vitro co-culture model, providing a theoretical basis for the clinical application of anti-CD20 monoclonal antibodies in TED.


Asunto(s)
Linfocitos B , Fibroblastos , Oftalmopatía de Graves , Receptor IGF Tipo 1 , Femenino , Humanos , Masculino , Linfocitos B/inmunología , Linfocitos B/metabolismo , Comunicación Celular , Células Cultivadas , Quimiocina CCL5/metabolismo , Técnicas de Cocultivo , Fibroblastos/metabolismo , Oftalmopatía de Graves/metabolismo , Oftalmopatía de Graves/inmunología , Interleucina-6/metabolismo , Depleción Linfocítica , Órbita/metabolismo , Órbita/inmunología , Receptor IGF Tipo 1/metabolismo , Rituximab/farmacología , Rituximab/uso terapéutico
2.
Retina ; 41(7): 1455-1462, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33332813

RESUMEN

PURPOSE: To determine associations of systemic medications with the incidence and growth of geographic atrophy (GA) in participants of the comparison of age-related macular degeneration treatments trials. METHODS: Participants of comparison of age-related macular degeneration treatments trials with new untreated choroidal neovascularization in the study eye (one study eye per participant) were randomized to receive treatment with bevacizumab or ranibizumab. Participants were released from clinical trial treatment at 2 years and examined at approximately 5 years. Color fundus photographs and fluorescein angiograms taken at baseline, Years 1, 2, and 5 were assessed for the presence and size of GA by two masked graders. Participants were interviewed about systemic medication use at baseline. Systemic medications previously reported to be associated with age-related macular degeneration were evaluated for associations with GA incidence in study eye using univariable and multivariable Cox models and for association with the GA growth using linear mixed effects models. RESULTS: In multivariable analysis of 1,011 study eyes without baseline GA, systemic medications, including cholinesterase inhibitors, angiotensin-converting enzyme inhibitors, calcium channel blockers, beta-blockers, diuretics, aspirin, steroids, statins, hormone replacement therapy, antacids, and drugs targeting G protein-coupled receptors, were not associated with GA incidence in the study eye (all adjusted hazard ratios ≤1.86, P ≥ 0.18). In multivariable analysis of 214 study eyes with longitudinal GA size measurements, calcium channel blockers were associated with a higher GA growth rate (0.40 vs. 0.30 mm/year, P = 0.02). CONCLUSION: None of the systemic medications analyzed were associated with GA incidence. However, calcium channel blockers were associated with a higher growth rate of GA in the study eye.


Asunto(s)
Bevacizumab/administración & dosificación , Atrofia Geográfica/tratamiento farmacológico , Degeneración Macular/tratamiento farmacológico , Ranibizumab/administración & dosificación , Agudeza Visual , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/administración & dosificación , Femenino , Angiografía con Fluoresceína/métodos , Estudios de Seguimiento , Fondo de Ojo , Atrofia Geográfica/diagnóstico , Atrofia Geográfica/epidemiología , Humanos , Incidencia , Inyecciones Intravítreas , Degeneración Macular/diagnóstico , Masculino , Factores de Tiempo , Tomografía de Coherencia Óptica/métodos , Resultado del Tratamiento , Estados Unidos/epidemiología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
3.
Am J Pathol ; 189(4): 826-838, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30711487

RESUMEN

Single-nucleotide polymorphisms and rare mutations in factor H (FH; official name, CFH) are associated with age-related macular degeneration and atypical hemolytic uremic syndrome, a form of thrombotic microangiopathy. Mice with the FH W1206R mutation (FHR/R) share features with human atypical hemolytic uremic syndrome. Herein, we report that FHR/R mice exhibited retinal vascular occlusion and ischemia. Retinal fluorescein angiography demonstrated delayed perfusion and vascular leakage in FHR/R mice. Optical coherence tomography imaging of FHR/R mice showed retinal degeneration, edema, and detachment. Histologic analysis of FHR/R mice revealed retinal thinning, vessel occlusion, as well as degeneration of photoreceptors and retinal pigment epithelium. Immunofluorescence showed albumin leakage from blood vessels into the neural retina, and electron microscopy demonstrated vascular endothelial cell irregularity with narrowing of retinal and choroidal vessels. Knockout of C6, a component of the membrane attack complex, prevented the aforementioned retinal phenotype in FHR/R mice, consistent with membrane attack complex-mediated pathogenesis. Pharmacologic blockade of C5 also rescued retinas of FHR/R mice. This FHR/R mouse strain represents a model for retinal vascular occlusive disorders and ischemic retinopathy. The results suggest complement dysregulation can contribute to retinal vascular occlusion and that an anti-C5 antibody might be helpful for C5-mediated thrombotic retinal diseases.


Asunto(s)
Factor H de Complemento/fisiología , Isquemia/etiología , Mutación , Neovascularización Patológica/etiología , Enfermedades de la Retina/etiología , Epitelio Pigmentado de la Retina/patología , Trombosis/etiología , Animales , Factor H de Complemento/genética , Isquemia/metabolismo , Isquemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Epitelio Pigmentado de la Retina/metabolismo , Trombosis/metabolismo , Trombosis/patología
4.
Kidney Int ; 96(1): 67-79, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30910380

RESUMEN

Atypical hemolytic uremic syndrome (aHUS) is a form of thrombotic microangiopathy (TMA) caused by dysregulated complement activation. Clinically, aHUS is effectively treated by an anti-C5 monoclonal antibody (mAb) but whether the disease is mediated by the C5a receptor (C5aR) or C5b-9 pathway, or both, is unknown. Here we address this in a factor H mutant mouse (FHR/R) which developed complement-mediated TMA as well as macrovascular thrombosis caused by an aHUS-related factor H point mutation (mouse W1206R, corresponding to human W1183R). C5 deficiency and anti-C5 mAb treatment blocked all disease manifestations in FHR/R mice. C5aR1 gene deficiency prevented macrovascular thrombosis in various organs but did not improve survival or reduce renal TMA. Conversely, C6 or C9 deficiency significantly improved survival and markedly diminished renal TMA but did not prevent macrovascular thrombosis. Interestingly, as they aged both FHR/R C6-/- and FHR/R C9-/- mice developed glomerular disease reminiscent of C3 glomerulonephritis. Thus, C5aR and C5b-9 pathways drove different aspects of disease in FHR/R mice with the C5aR pathway being responsible for macrovascular thrombosis and chronic inflammatory injury while the C5b-9 pathway caused renal TMA. Our data provide new understanding of the pathogenesis of complement-mediated TMA and macrovascular thrombosis in FHR/R mice and suggest that C5 blockade is more effective for the treatment of aHUS than selectively targeting the C5aR or C5b-9 pathway alone.


Asunto(s)
Síndrome Hemolítico Urémico Atípico/inmunología , Factor H de Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Glomérulos Renales/patología , Receptor de Anafilatoxina C5a/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/genética , Síndrome Hemolítico Urémico Atípico/patología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/genética , Activación de Complemento/inmunología , Complemento C6/genética , Complemento C6/inmunología , Complemento C6/metabolismo , Factor H de Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Glomérulos Renales/irrigación sanguínea , Glomérulos Renales/ultraestructura , Masculino , Ratones , Ratones Transgénicos , Microscopía Electrónica , Mutación Puntual , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/metabolismo
5.
Blood ; 129(9): 1184-1196, 2017 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-28057640

RESUMEN

Complement plays a key role in host defense, but its dysregulation can cause autologous tissue injury. Complement activation is normally controlled by regulatory proteins, including factor H (FH) in plasma and membrane cofactor protein (MCP) on the cell surface. Mutations in FH and MCP are linked to atypical hemolytic uremic syndrome, a type of thrombotic microangiopathy (TMA) that causes renal failure. We describe here that disruption of FH function on the cell surface can also lead to disseminated complement-dependent macrovascular thrombosis. By gene targeting, we introduced a point mutation (W1206R) into murine FH that impaired its interaction with host cells but did not affect its plasma complement-regulating activity. Homozygous mutant mice carrying this mutation developed renal TMA as well as systemic thrombophilia involving large blood vessels in multiple organs, including liver, lung, spleen, and kidney. Approximately 30% of mutant mice displayed symptoms of stroke and ischemic retinopathy, and 48% died prematurely. Genetic deficiency of complement C3 and factor D prevented both the systemic thrombophilia and renal TMA phenotypes. These results demonstrate a causal relationship between complement dysregulation and systemic angiopathy and suggest that complement activation may contribute to various human thrombotic disorders involving both the micro- and macrovasculature.


Asunto(s)
Factor H de Complemento/genética , Síndrome Hemolítico-Urémico/genética , Trombofilia/genética , Animales , Western Blotting , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Puntual
6.
Retina ; 39(10): 1965-1972, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30157115

RESUMEN

PURPOSE: Because patients often take iron supplements without medical indication, and iron can accumulate in vascular endothelial cells, the authors evaluated the association of oral iron supplementation with retinal/subretinal hemorrhage in patients with neovascular age-related macular degeneration. METHODS: A post hoc secondary data analysis of comparison of age-related macular degeneration treatments trials was performed. Participants were interviewed for use of oral iron supplements. Trained readers evaluated retinal/subretinal hemorrhage in baseline fundus photographs. Adjusted odds ratios from multivariate logistic regression models assessed the association between iron use and baseline hemorrhage adjusted by age, sex, smoking, hypertension, anemia, and use of antiplatelet/anticoagulant drugs. RESULTS: Among 1,165 participants, baseline retinal/subretinal hemorrhage was present in the study eye in 71% of 181 iron users and in 61% of 984 participants without iron use (adjusted odds ratio = 1.47, P = 0.04), and the association was dose dependent (adjusted linear trend P = 0.048). Iron use was associated with hemorrhage in participants with hypertension (adjusted odds ratio = 1.87, P = 0.006) but not without hypertension. The association of iron use with hemorrhage remained significant among hypertensive participants without anemia (adjusted odds ratio = 1.85, P = 0.02). CONCLUSION: Among participants of comparison of age-related macular degeneration treatments trials, the use of oral iron supplements was associated with retinal/subretinal hemorrhage in a dose-response manner. Unindicated iron supplementation may be detrimental in patients with wet age-related macular degeneration.


Asunto(s)
Compuestos de Hierro/efectos adversos , Ranibizumab/administración & dosificación , Hemorragia Retiniana/inducido químicamente , Agudeza Visual , Degeneración Macular Húmeda/tratamiento farmacológico , Administración Oral , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis , Suplementos Dietéticos , Femenino , Angiografía con Fluoresceína , Fondo de Ojo , Humanos , Inyecciones Intravítreas , Compuestos de Hierro/administración & dosificación , Masculino , Hemorragia Retiniana/diagnóstico , Tomografía de Coherencia Óptica , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Degeneración Macular Húmeda/diagnóstico
7.
Retina ; 39(3): 502-513, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29324592

RESUMEN

PURPOSE: To assess the potential ocular toxicity of a combined BRAF inhibition (BRAFi) + MEK inhibition (MEKi) + hydroxychloroquine (HCQ) regime used to treat metastatic BRAF mutant melanoma. METHODS: Patients with stage IV metastatic melanoma and BRAF V600E mutations (n = 11, 31-68 years of age) were included. Treatment was with oral dabrafenib, 150 mg bid, trametinib, 2 mg/day, and HCQ, 400 mg to 600 mg bid. An ophthalmic examination, spectral domain optical coherence tomography, near-infrared and short-wavelength fundus autofluorescence, and static perimetry were performed at baseline, 1 month, and q/6 months after treatment. RESULTS: There were no clinically significant ocular events; there was no ocular inflammation. The only medication-related change was a separation of the photoreceptor outer segment tip from the apical retinal pigment epithelium that could be traced from the fovea to the perifoveal retina noted in 9/11 (82%) of the patients. There were no changes in retinal pigment epithelium melanization or lipofuscin content by near-infrared fundus autofluorescence and short-wavelength fundus autofluorescence, respectively. There were no inner retinal or outer nuclear layer changes. Visual acuities and sensitivities were unchanged. CONCLUSION: BRAFi (trametinib) + MEKi (dabrafenib) + HCQ causes very frequent, subclinical separation of the photoreceptor outer segment from the apical retinal pigment epithelium without inner retinal changes or signs of inflammation. The changes suggest interference with the maintenance of the outer retinal barrier and/or phagocytic/pump functions of the retinal pigment epithelium by effective MEK inhibition.


Asunto(s)
Antineoplásicos/efectos adversos , Inhibidores Enzimáticos/efectos adversos , Hidroxicloroquina/efectos adversos , Imidazoles/efectos adversos , Mácula Lútea/patología , Melanoma/tratamiento farmacológico , Oximas/efectos adversos , Inhibidores de Proteínas Quinasas/efectos adversos , Piridonas/efectos adversos , Pirimidinonas/efectos adversos , Enfermedades de la Retina , Adulto , Anciano , Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos , Hidroxicloroquina/uso terapéutico , Imidazoles/uso terapéutico , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Masculino , Melanoma/genética , Persona de Mediana Edad , Oximas/uso terapéutico , Células Fotorreceptoras/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Piridonas/uso terapéutico , Pirimidinonas/uso terapéutico , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/patología , Epitelio Pigmentado de la Retina/patología
8.
Mol Vis ; 23: 171-178, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458506

RESUMEN

PURPOSE: (-)-epigallocatechin-3-gallate (EGCG), a major catechin component of green tea, is reported to delay or prevent certain forms of cancer, arthritis, cardiovascular disease, and neurodegenerative disorders. In this study, we determined if systemically administered EGCG could protect the retina against light damage (LD) in mice. METHODS: BALB/cJ mice were treated with either EGCG or saline via intraperitoneal (IP) injection, and then placed under constant cool white light-emitting diode (LED) light (10,000 lux) for 5 h. Retinal structure and function were evaluated using optical coherence tomography (OCT), histology, and electroretinography (ERG) 7 days after LD. In addition, the mRNAs of several oxidative stress genes were quantified by qPCR before LD and 24 h after LD. RESULTS: OCT and photomicrographs of mouse retinas showed morphologic protection of photoreceptors. Mice in the EGCG group had significantly higher ERG amplitudes for all three wave types compared with mice in the saline control group, which indicated that EGCG protected retinal function. Furthermore, qPCR results showed that EGCG administration can increase the mRNA level of the antioxidant gene Sod2 before LD and 24 h after LD. CONCLUSIONS: The IP injection of EGCG attenuated the detrimental effects of bright light on the retinas of BALB/cJ mice by protecting the structure and function of the retina.


Asunto(s)
Antioxidantes/uso terapéutico , Catequina/análogos & derivados , Luz/efectos adversos , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Traumatismos Experimentales por Radiación/prevención & control , Degeneración Retiniana/prevención & control , Animales , Antioxidantes/administración & dosificación , Catequina/administración & dosificación , Catequina/uso terapéutico , Electrorretinografía , Inyecciones Intraperitoneales , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo/genética , ARN Mensajero/genética , Traumatismos Experimentales por Radiación/etiología , Traumatismos Experimentales por Radiación/fisiopatología , Reacción en Cadena en Tiempo Real de la Polimerasa , Retina/fisiopatología , Degeneración Retiniana/etiología , Degeneración Retiniana/fisiopatología , Superóxido Dismutasa/genética , Tomografía de Coherencia Óptica
9.
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
10.
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
11.
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
12.
J Biol Chem ; 290(19): 11918-34, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25802332

RESUMEN

Dysregulation of iron homeostasis may be a pathogenic factor in age-related macular degeneration (AMD). Meanwhile, the formation of complement-containing deposits under the retinal pigment epithelial (RPE) cell layer is a pathognomonic feature of AMD. In this study, we investigated the molecular mechanisms by which complement component 3 (C3), a central protein in the complement cascade, is up-regulated by iron in RPE cells. Modulation of TGF-ß signaling, involving ERK1/2, SMAD3, and CCAAT/enhancer-binding protein-δ, is responsible for iron-induced C3 expression. The differential effects of spatially distinct SMAD3 phosphorylation sites at the linker region and at the C terminus determined the up-regulation of C3. Pharmacologic inhibition of either ERK1/2 or SMAD3 phosphorylation decreased iron-induced C3 expression levels. Knockdown of SMAD3 blocked the iron-induced up-regulation and nuclear accumulation of CCAAT/enhancer-binding protein-δ, a transcription factor that has been shown previously to bind the basic leucine zipper 1 domain in the C3 promoter. We show herein that mutation of this domain reduced iron-induced C3 promoter activity. In vivo studies support our in vitro finding of iron-induced C3 up-regulation. Mice with a mosaic pattern of RPE-specific iron overload demonstrated co-localization of iron-induced ferritin and C3d deposits. Humans with aceruloplasminemia causing RPE iron overload had increased RPE C3d deposition. The molecular events in the iron-C3 pathway represent therapeutic targets for AMD or other diseases exacerbated by iron-induced local complement dysregulation.


Asunto(s)
Complemento C3/metabolismo , Degeneración Macular/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Línea Celular , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Hierro/química , Sobrecarga de Hierro , Ligandos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Transducción de Señal , Proteína smad3/metabolismo , Regulación hacia Arriba
13.
Exp Eye Res ; 145: 1-9, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26475979

RESUMEN

Oxidative stress and inflammation play key roles in the light damage (LD) model of photoreceptor degeneration, as well as in age-related macular degeneration (AMD). We sought to investigate whether Berberine (BBR), an antioxidant herb extract, would protect the retina against light-induced degeneration. To accomplish this, Balb/c mice were treated with BBR or PBS via gavage for 7 days, and then were placed in constant cool white light-emitting diode (LED) light (10,000 lux) for 4 h. Retinal function and degeneration were evaluated by histology, electroretinography (ERG) and optical coherence tomography (OCT) at 7d after LD. Additionally, mRNA levels of cell-type specific, antioxidant, and inflammatory genes were compared 7d after LD. Photoreceptor DNA fragmentation was assessed via the terminal deoxynucleotidyl transferase dUTP nick end-labeling (TUNEL) assay. LD resulted in substantial photoreceptor-specific cell death. Histological analysis using plastic sections showed dosing with BBR preserved photoreceptors. The ERG analysis demonstrated functional protection by BBR in rod-b, -a, and cone-b waves. In OCT images, mice receiving PBS showed severe thinning and disorganization of the photoreceptor layer 7 days after LD, whereas mice treated with BBR had significantly less thinning and disorganization. Consistent with OCT results, the mRNA levels of Rho in the NSR, and Rpe65 and Mct3 in the RPE, were significantly higher in mice treated with BBR. The numbers of TUNEL-positive photoreceptors were significantly decreased in BBR-treated mice. The retinal mRNA levels of oxidative stress genes, the number of microglia/macrophages, and the malondialdehyde (MDA) immunolabeling were significantly lower in BBR-treated mice compared to controls 48 h after LD, which indicates oxidative stress was reduced by BBR in light-damaged eyes. In conclusion, systemic BBR is protective against light-induced retinal degeneration associated with diminished oxidative stress in the retina. These results suggest that BBR may be protective against retinal diseases involving oxidative stress.


Asunto(s)
Berberina/farmacología , Luz/efectos adversos , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Degeneración Retiniana/prevención & control , Animales , Muerte Celular , Modelos Animales de Enfermedad , Electrorretinografía , Etiquetado Corte-Fin in Situ , Masculino , Ratones , Ratones Endogámicos BALB C , Estrés Oxidativo , Células Fotorreceptoras de Vertebrados/patología , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Degeneración Retiniana/etiología , Degeneración Retiniana/patología , Rodopsina/metabolismo , Tomografía de Coherencia Óptica
14.
Exp Eye Res ; 151: 122-33, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27565570

RESUMEN

Iron accumulation in the retina is associated with the development of age-related macular degeneration (AMD). IV iron is a common method to treat iron deficiency anemia in adults, and its retinal manifestations have not hitherto been identified. To assess whether IV iron formulations can be retina-toxic, we generated a mouse model for iron-induced retinal damage. Male C57BL/6J mice were randomized into groups receiving IV iron-sucrose (+Fe) or 30% sucrose (-Fe). Iron levels in neurosensory retina (NSR), retinal pigment epithelium (RPE), and choroid were assessed using immunofluorescence, quantitative PCR, and the Perls' iron stain. Iron levels were most increased in the RPE and choroid while levels in the NSR did not differ significantly in +Fe mice compared to controls. Eyes from +Fe mice shared histological features with AMD, including Bruch's membrane (BrM) thickening with complement C3 deposition, as well as RPE hypertrophy and vacuolization. This focal degeneration correlated with areas of high choroidal iron levels. Ultrastructural analysis provided further detail of the RPE/photoreceptor outer segment vacuolization and Bruch's membrane thickening. Findings were correlated with a clinical case of a 43-year-old patient who developed numerous retinal drusen, the hallmark of AMD, within 11 months of IV iron therapy. Our results suggest that IV iron therapy may have the potential to induce or exacerbate a form of retinal degeneration. This retinal degeneration shares features with AMD, indicating the need for further study of AMD risk in patients receiving IV iron treatment.


Asunto(s)
Compuestos Férricos/efectos adversos , Ácido Glucárico/efectos adversos , Hierro/metabolismo , Degeneración Macular/inducido químicamente , Estrés Oxidativo/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Animales , Apoferritinas/biosíntesis , Apoferritinas/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Compuestos Férricos/administración & dosificación , Sacarato de Óxido Férrico , Regulación de la Expresión Génica , Ácido Glucárico/administración & dosificación , Inyecciones Intravenosas , Degeneración Macular/genética , Degeneración Macular/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Transferrina/biosíntesis , Receptores de Transferrina/genética , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo
15.
Am J Pathol ; 184(11): 2862-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25174877

RESUMEN

The retina can be shielded by the blood-retinal barrier. Because photoreceptors are damaged by excess iron, it is important to understand whether the blood-retinal barrier protects against high serum iron levels. Bone morphogenic protein 6 (Bmp6) knockout mice have serum iron overload. Herein, we tested whether the previously documented retinal iron accumulation in Bmp6 knockout mice might result from the high serum iron levels or, alternatively, low levels of retinal hepcidin, an iron regulatory hormone whose transcription can be up-regulated by Bmp6. Furthermore, to determine whether increases in serum iron can elevate retinal iron levels, we i.v. injected iron into wild-type mice. Retinas were analyzed by real-time quantitative PCR and immunofluorescence to assess the levels of iron-regulated genes/proteins and oxidative stress. Retinal hepcidin mRNA levels in Bmp6 knockout retinas were the same as, or greater than, those in age-matched wild-type retinas, indicating that Bmp6 knockout does not cause retinal hepcidin deficiency. Changes in mRNA levels of L ferritin and transferrin receptor indicated increased retinal iron levels in i.v. iron-injected wild-type mice. Oxidative stress markers were elevated in photoreceptors of mice receiving i.v. iron. These findings suggest that elevated serum iron levels can overwhelm local retinal iron regulatory mechanisms.


Asunto(s)
Barrera Hematorretinal/metabolismo , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Retina/metabolismo , Animales , Proteína Morfogenética Ósea 6/genética , Proteína Morfogenética Ósea 6/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Hierro/sangre , Sobrecarga de Hierro/genética , Ratones , Ratones Noqueados , Estrés Oxidativo/fisiología
16.
Am J Pathol ; 184(10): 2721-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25088982

RESUMEN

Mutations in the RP1 gene can cause retinitis pigmentosa. We identified a spontaneous L66P mutation caused by two adjacent point mutations in the Rp1 gene in a colony of C57BL/6J mice. Mice homozygous for the L66P mutation exhibited slow, progressive photoreceptor degeneration throughout their lifespan. Optical coherence tomography imaging found abnormal photoreceptor reflectivity at 1 month of age. Histology found shortening and disorganization of the photoreceptor inner and outer segments and progressive thinning of the outer nuclear layer. Electroretinogram a- and b-wave amplitudes were decreased with age. Western blot analysis found that the quantity and size of the mutated retinitis pigmentosa 1 (RP1) protein were normal. However, immunohistochemistry found that the mutant Rp1 protein partially mislocalized to the transition zone of the shortened axonemes. This mutation disrupted colocalization with cytoplasmic microtubules in vitro. In conclusion, the L66P mutation in the first doublecortin domain of the Rp1 gene impairs Rp1 protein localization and function, leading to abnormalities in photoreceptor outer segment structure and progressive photoreceptor degeneration. This is the first missense mutation in Rp1 shown to cause retinal degeneration. It provides a unique, slowly progressive photoreceptor degeneration model that mirrors the slow degeneration kinetics in most patients with retinitis pigmentosa.


Asunto(s)
Axonema/metabolismo , Proteínas del Ojo/genética , Proteínas Asociadas a Microtúbulos/genética , Degeneración Retiniana/genética , Animales , Células COS , Chlorocebus aethiops , Electrorretinografía , Proteínas del Ojo/metabolismo , Femenino , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Mutación Missense , Células Fotorreceptoras/metabolismo , Degeneración Retiniana/metabolismo
17.
Exp Eye Res ; 128: 92-101, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25277027

RESUMEN

The purpose of our studies was to examine the relationship between iron and melanogenesis in retinal pigment epithelial cells, as prior observations had suggested that iron may promote melanogenesis. This relationship has potential clinical importance, as both iron overload and hyperpigmentation are associated with age-related macular degeneration (AMD). Human fetal retinal pigment epithelial cells and ARPE-19 cells were treated with iron in the form of ferric ammonium citrate, after which quantitative RT-PCR and electron microscopy were performed. Melanogenesis genes tyrosinase, tyrosinase-related protein 1, Hermansky-Pudlak Syndrome 3, premelanosome protein and dopachrome tautomerase were upregulated, as was the melanogenesis-controlling transcription factor, microphthalmia-associated transcription factor (MITF). Iron-treated cells had increased pigmentation and melanosome number. Multiple transcription factors upstream of MITF were upregulated by iron.


Asunto(s)
Compuestos Férricos/farmacología , Melaninas/biosíntesis , Melanosomas/metabolismo , Compuestos de Amonio Cuaternario/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Regulación hacia Arriba/fisiología , Western Blotting , Proteínas Portadoras/genética , Células Cultivadas , Edad Gestacional , Humanos , Péptidos y Proteínas de Señalización Intracelular , Oxidorreductasas Intramoleculares/genética , Glicoproteínas de Membrana/genética , Monofenol Monooxigenasa/genética , Oxidorreductasas/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Epitelio Pigmentado de la Retina/embriología , Epitelio Pigmentado de la Retina/metabolismo , Donantes de Tejidos , Antígeno gp100 del Melanoma/genética
18.
Am J Pathol ; 180(4): 1614-24, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22342521

RESUMEN

Hephaestin (Heph) is a ferroxidase protein that converts ferrous to ferric iron to facilitate cellular iron export by ferroportin. Many tissues express either Heph or its homologue, ceruloplasmin (Cp), but the retina expresses both. In mice, a combined systemic mutation of Heph and systemic knockout of Cp (Cp(-/-), Heph(sla/sla)) causes retinal iron accumulation and retinal degeneration, with features of human age-related macular degeneration; however, the role of Heph and Cp in the individual retinal cells is unclear. Herein, we used conditional knockout mice to study Heph's role in retinal pigment epithelial (RPE) and photoreceptor cells. Loss of both Heph and Cp from RPE cells alone results in RPE cell iron accumulation and degeneration. We found, however, that RPE iron accumulation in these conditional knockout mice is not as great as in systemic knockout mice. Photoreceptor-specific Heph knockout indicates that the additional iron in the RPE cells does not result from loss of ferroxidases in the photoreceptors, and Cp and Heph play minor roles in photoreceptors. Instead, loss of ferroxidases in other retinal cells causes retinal iron accumulation and transfer of iron to the RPE cells. Cp and Heph are necessary for iron export from the retina but are not essential for iron import into the retina. Thus, our studies, revise how we think about iron import and export from the retina.


Asunto(s)
Degeneración Macular/metabolismo , Proteínas de la Membrana/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Células Cultivadas , Ceruloplasmina/metabolismo , Modelos Animales de Enfermedad , Hierro/metabolismo , Degeneración Macular/genética , Degeneración Macular/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Noqueados , Mutación , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Neuronas Retinianas/metabolismo , Epitelio Pigmentado de la Retina/patología
19.
Retin Cases Brief Rep ; 17(4): 340-342, 2023 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34618713

RESUMEN

PURPOSE: To report a novel presentation of bilateral paracentral acute middle maculopathy and peripheral vascular occlusions in Behcet disease. METHODS: A retrospective case report with multimodal imaging studies of a patient with Behcet's disease. RESULTS: A 58-year-old Chinese man presented with a paracentral scotoma, fever, arthralgias, and skin rash. Human leukocyte antigen (HLA) typing revealed HLA-B51 positivity. Ophthalmic examination showed peripheral retinal hemorrhages and fluorescein angiography (FA) demonstrated vascular occlusions in the peripheral retina bilaterally. Optical coherence tomography showed classic acute paracentral acute middle maculopathy lesions in both eyes. CONCLUSIONS: Paracentral acute middle maculopathy and peripheral vascular occlusion are infrequent and unconventional presentations of Behcet disease. To the best of our knowledge, this is the first report in the ophthalmic literature of paracentral acute middle maculopathy and peripheral vascular occlusion in Behcet disease.


Asunto(s)
Síndrome de Behçet , Degeneración Macular , Enfermedades de la Retina , Masculino , Humanos , Persona de Mediana Edad , Síndrome de Behçet/complicaciones , Síndrome de Behçet/diagnóstico , Estudios Retrospectivos , Enfermedad Aguda , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/etiología , Enfermedades de la Retina/patología , Angiografía con Fluoresceína/métodos , Tomografía de Coherencia Óptica/métodos , Degeneración Macular/etiología , Degeneración Macular/complicaciones , Vasos Retinianos/patología
20.
Artículo en Inglés | MEDLINE | ID: mdl-38055904

RESUMEN

PURPOSE: To report the case of severe bilateral retinal vascular occlusion in a patient with hyperoxalosis and chronic renal failure. METHODS: Observational case report. Medical and imaging records were retrospectively reviewed. The patient was imaged with ultra-widefield (UWF) fundus photography and fluorescein angiography (UWF-FA), cross sectional and en face spectral-domain optical coherence tomography (SD-OCT), and OCT angiography. RESULTS: A 32-year-old diabetic patient receiving peritoneal dialysis was referred because of severe vision loss. UWF color fundus photography showed diffuse sclerotic retinal vessels and diffuse intraretinal crystals in both eyes. UWF-FA illustrated near-complete retinal vascular occlusion and capillary wipe out in both eyes. SD-OCT demonstrated diffuse inner and middle retina thinning in both eyes and multiple intraretinal hyperreflective foci consistent with crystalline deposits in all retina layers of both eyes. OCT angiography revealed severe capillary and large vessel non-perfusion in the superficial and deep retinal capillary plexus of each eye. The serum oxalate levels were increased at 28 µmol/L (reference range < 2 µmol/L) and genetic testing was positive for a heterozygous mutation of the AGXT (Alanine-Glyoxylate Amino Transferase) gene that causes type 1 autosomal recessive primary hyperoxaluria. CONCLUSION: A diagnosis of hyperoxalosis causing severe retinal vascular occlusion was rendered. Hyperoxalosis was the result of multiple factors including heterozygous AGXT mutation, chronic renal failure insufficiently treated with peritoneal dialysis, and a diet high in oxalate. This case highlights the importance of ruling out retinal oxalosis in patients on peritoneal dialysis in order to initiate prompt hemodialysis and prevent severe retinal vascular occlusion.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA