Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
2.
Proc Natl Acad Sci U S A ; 112(39): 12163-8, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26374840

RESUMEN

Pathologic ocular neovascularization commonly causes blindness. It is critical to identify the factors altered in pathologically proliferating versus normally quiescent vessels to develop effective targeted therapeutics. MicroRNAs regulate both physiological and pathological angiogenesis through modulating expression of gene targets at the posttranscriptional level. However, it is not completely understood if specific microRNAs are altered in pathologic ocular blood vessels, influencing vascular eye diseases. Here we investigated the potential role of a specific microRNA, miR-150, in regulating ocular neovascularization. We found that miR-150 was highly expressed in normal quiescent retinal blood vessels and significantly suppressed in pathologic neovessels in a mouse model of oxygen-induced proliferative retinopathy. MiR-150 substantially decreased endothelial cell function including cell proliferation, migration, and tubular formation and specifically suppressed the expression of multiple angiogenic regulators, CXCR4, DLL4, and FZD4, in endothelial cells. Intravitreal injection of miR-150 mimic significantly decreased pathologic retinal neovascularization in vivo in both wild-type and miR-150 knockout mice. Loss of miR-150 significantly promoted angiogenesis in aortic rings and choroidal explants ex vivo and laser-induced choroidal neovascularization in vivo. In conclusion, miR-150 is specifically enriched in quiescent normal vessels and functions as an endothelium-specific endogenous inhibitor of pathologic ocular neovascularization.


Asunto(s)
Neovascularización Coroidal/genética , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/metabolismo , MicroARNs/farmacología , Neovascularización Retiniana/genética , Vasos Retinianos/citología , Regiones no Traducidas 3'/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Proteínas de Unión al Calcio , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neovascularización Coroidal/metabolismo , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Inyecciones Intravítreas , Captura por Microdisección con Láser , Luciferasas , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , MicroARNs/administración & dosificación , MicroARNs/genética , Datos de Secuencia Molecular , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/metabolismo
3.
BMC Biol ; 15(1): 113, 2017 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-29183319

RESUMEN

BACKGROUND: Peroxisome proliferator activated receptor-alpha (PPARα) is a ubiquitously expressed nuclear receptor. The role of endogenous PPARα in retinal neuronal homeostasis is unknown. Retinal photoreceptors are the highest energy-consuming cells in the body, requiring abundant energy substrates. PPARα is a known regulator of lipid metabolism, and we hypothesized that it may regulate lipid use for oxidative phosphorylation in energetically demanding retinal neurons. RESULTS: We found that endogenous PPARα is essential for the maintenance and survival of retinal neurons, with Pparα -/- mice developing retinal degeneration first detected at 8 weeks of age. Using extracellular flux analysis, we identified that PPARα mediates retinal utilization of lipids as an energy substrate, and that ablation of PPARα ultimately results in retinal bioenergetic deficiency and neurodegeneration. This may be due to PPARα regulation of lipid transporters, which facilitate the internalization of fatty acids into cell membranes and mitochondria for oxidation and ATP production. CONCLUSION: We identify an endogenous role for PPARα in retinal neuronal survival and lipid metabolism, and furthermore underscore the importance of fatty acid oxidation in photoreceptor survival. We also suggest PPARα as a putative therapeutic target for age-related macular degeneration, which may be due in part to decreased mitochondrial efficiency and subsequent energetic deficits.


Asunto(s)
Ácidos Grasos/metabolismo , Metabolismo de los Lípidos , PPAR alfa/genética , Retina/metabolismo , Neuronas Retinianas/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción , PPAR alfa/metabolismo , Ratas , Ratas Sprague-Dawley
5.
Am J Pathol ; 186(10): 2588-600, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27524797

RESUMEN

Familial exudative vitreoretinopathy (FEVR) is characterized by delayed retinal vascular development, which promotes hypoxia-induced pathologic vessels. In severe cases FEVR may lead to retinal detachment and visual impairment. Genetic studies linked FEVR with mutations in Wnt signaling ligand or receptors, including low-density lipoprotein receptor-related protein 5 (LRP5) gene. Here, we investigated ocular pathologies in a Lrp5 knockout (Lrp5(-/-)) mouse model of FEVR and explored whether treatment with a pharmacologic Wnt activator lithium could bypass the genetic defects, thereby protecting against eye pathologies. Lrp5(-/-) mice displayed significantly delayed retinal vascular development, absence of deep layer retinal vessels, leading to increased levels of vascular endothelial growth factor and subsequent pathologic glomeruloid vessels, as well as decreased inner retinal visual function. Lithium treatment in Lrp5(-/-) mice significantly restored the delayed development of retinal vasculature and the intralaminar capillary networks, suppressed formation of pathologic glomeruloid structures, and promoted hyaloid vessel regression. Moreover, lithium treatment partially rescued inner-retinal visual function and increased retinal thickness. These protective effects of lithium were largely mediated through restoration of canonical Wnt signaling in Lrp5(-/-) retina. Lithium treatment also substantially increased vascular tubular formation in LRP5-deficient endothelial cells. These findings suggest that pharmacologic activation of Wnt signaling may help treat ocular pathologies in FEVR and potentially other defective Wnt signaling-related diseases.


Asunto(s)
Litio/farmacología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Enfermedades de la Retina/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Enfermedades Hereditarias del Ojo , Vitreorretinopatías Exudativas Familiares , Femenino , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Retina/efectos de los fármacos , Retina/embriología , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/embriología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 36(9): 1919-27, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417579

RESUMEN

OBJECTIVE: Pathological ocular neovascularization is a major cause of blindness. Increased dietary intake of ω-3 long-chain polyunsaturated fatty acids (LCPUFA) reduces retinal neovascularization and choroidal neovascularization (CNV), but ω-3 LCPUFA metabolites of a major metabolizing pathway, cytochrome P450 oxidase (CYP) 2C, promote ocular pathological angiogenesis. We hypothesized that inhibition of CYP2C activity will add to the protective effects of ω-3 LCPUFA on neovascular eye diseases. APPROACH AND RESULTS: The mouse models of oxygen-induced retinopathy and laser-induced CNV were used to investigate pathological angiogenesis in the retina and choroid, respectively. The plasma levels of ω-3 LCPUFA metabolites of CYP2C were determined by mass spectroscopy. Aortic ring and choroidal explant sprouting assays were used to investigate the effects of CYP2C inhibition and ω-3 LCPUFA-derived CYP2C metabolic products on angiogenesis ex vivo. We found that inhibition of CYP2C activity by montelukast added to the protective effects of ω-3 LCPUFA on retinal neovascularization and CNV by 30% and 20%, respectively. In CYP2C8-overexpressing mice fed a ω-3 LCPUFA diet, montelukast suppressed retinal neovascularization and CNV by 36% and 39% and reduced the plasma levels of CYP2C8 products. Soluble epoxide hydrolase inhibition, which blocks breakdown and inactivation of CYP2C ω-3 LCPUFA-derived active metabolites, increased oxygen-induced retinopathy and CNV in vivo. Exposure to selected ω-3 LCPUFA metabolites of CYP2C significantly reversed the suppression of both angiogenesis ex vivo and endothelial cell functions in vitro by the CYP2C inhibitor montelukast. CONCLUSIONS: Inhibition of CYP2C activity adds to the protective effects of ω-3 LCPUFA on pathological retinal neovascularization and CNV.


Asunto(s)
Acetatos/farmacología , Inhibidores de la Angiogénesis/farmacología , Neovascularización Coroidal/prevención & control , Inhibidores del Citocromo P-450 CYP2C8/farmacología , Citocromo P-450 CYP2C8/metabolismo , Ácidos Grasos Omega-3/farmacología , Quinolinas/farmacología , Neovascularización Retiniana/prevención & control , Retinopatía de la Prematuridad/prevención & control , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Células Cultivadas , Neovascularización Coroidal/enzimología , Neovascularización Coroidal/genética , Neovascularización Coroidal/fisiopatología , Ciclopropanos , Citocromo P-450 CYP2C8/genética , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Ácidos Grasos Omega-3/metabolismo , Genotipo , Humanos , Hiperoxia/complicaciones , Rayos Láser , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Fenotipo , Neovascularización Retiniana/enzimología , Neovascularización Retiniana/genética , Neovascularización Retiniana/fisiopatología , Retinopatía de la Prematuridad/enzimología , Retinopatía de la Prematuridad/genética , Retinopatía de la Prematuridad/fisiopatología , Sulfuros , Técnicas de Cultivo de Tejidos
7.
J Crohns Colitis ; 17(4): 504-512, 2023 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-36272109

RESUMEN

BACKGROUND: Upadacitinib is a selective Janus kinase inhibitor approved for the management of ulcerative colitis and is under evaluation for the management of Crohn's disease [CD] in Phase 3 clinical trials. AIMS: Our goal was to describe our real-world experience with upadacitinib in CD. METHODS: This is a two-centre retrospective cohort study of adult patients with moderate to severe CD on upadacitinib. The primary outcome was clinical response and remission as determined by stool frequency and abdominal pain scores. Secondary endpoints included endoscopic response and remission as determined by change in Simple Endoscopic Score for CD. Outcomes were assessed at 3 months after starting upadacitinib and at the patient's most recent follow-up. We further evaluated adverse events and dose-related response. RESULTS: A total of 45 CD patients received upadacitinib and were included in the safety analysis. Thirty-six patients received upadacitinib for CD, whereas nine received it for inflammatory arthritis [n = 8] or pyoderma [n = 1]. Thirty-three patients received upadacitinib for 3 months or longer and were included in the efficacy analysis. At the 3-month follow-up, 21 patients achieved clinical response [63.6%] and nine achieved clinical remission [27.2%]. At time of last follow-up, 23 patients had clinical response [69.7%], ten achieved clinical remission [30.3%] and four [28.6%] achieved endoscopic remission. Adverse events occurred in 12 patients [26.7%]. Two patients had a serious adverse event [4.5%] without associated mortality. CONCLUSION: In this real-world cohort of highly refractory CD patients, upadacitinib was effective in inducing remission and had an acceptable safety profile.


Asunto(s)
Enfermedad de Crohn , Compuestos Heterocíclicos con 3 Anillos , Inhibidores de las Cinasas Janus , Enfermedad de Crohn/tratamiento farmacológico , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Inhibidores de las Cinasas Janus/uso terapéutico , Estudios Retrospectivos , Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Resultado del Tratamiento
8.
Crohns Colitis 360 ; 4(2): otac008, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36777041

RESUMEN

Background: Microscopic colitis (MC) is suspected to result from increased immune activity in gut mucosa. Immune checkpoint inhibitors (ICIs) treat cancer by activating the immune system, and further investigation is needed regarding their role in the development of MC. Methods: A retrospective case series investigated cases of endoscopically and histologically confirmed MC developing after administration of ICIs. Clinical notes and medication administration records were reviewed for demographics, symptom duration, and treatment response. Results: Nineteen cases of de novo MC were identified, with 95% of cases requiring steroid treatment, 53% presenting with hospitalization, and colitis-related mortality in 1 individual. Symptom onset occurred a median of 160 days after initiation of ICI therapy and 53 days after their most recent dose of therapy. Patients had a median of 125 days of symptoms, and ICI therapy was held in 70% of individuals due for treatment. Conclusions: MC can develop after ICI administration, and presents with severe symptoms, often requiring hospitalization and steroid treatment. In certain individuals this can require a prolonged treatment course of steroid therapy or immunomodulators. Individuals developing diarrhea after ICI therapy warrant thorough workup including endoscopy and rapid treatment initiation given the disease severity observed in this series.

10.
Cell Rep ; 18(7): 1606-1613, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28199833

RESUMEN

Pathological neovascularization, a leading cause of blindness, is seen in retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration. Using a mouse model of hypoxia-driven retinal neovascularization, we find that fibroblast growth factor 21 (FGF21) administration suppresses, and FGF21 deficiency worsens, retinal neovessel growth. The protective effect of FGF21 against neovessel growth was abolished in adiponectin (APN)-deficient mice. FGF21 administration also decreased neovascular lesions in two models of neovascular age-related macular degeneration: very-low-density lipoprotein-receptor-deficient mice with retinal angiomatous proliferation and laser-induced choroidal neovascularization. FGF21 inhibited tumor necrosis α (TNF-α) expression but did not alter Vegfa expression in neovascular eyes. These data suggest that FGF21 may be a therapeutic target for pathologic vessel growth in patients with neovascular eye diseases, including retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration.


Asunto(s)
Neovascularización Coroidal/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/farmacología , Neovascularización Patológica/tratamiento farmacológico , Retina/efectos de los fármacos , Neovascularización Retiniana/tratamiento farmacológico , Animales , Neovascularización Coroidal/metabolismo , Modelos Animales de Enfermedad , Degeneración Macular/tratamiento farmacológico , Degeneración Macular/metabolismo , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Retina/metabolismo , Neovascularización Retiniana/metabolismo , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
J Exp Med ; 214(6): 1753-1767, 2017 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-28465464

RESUMEN

Pathological neovessels growing into the normally avascular photoreceptors cause vision loss in many eye diseases, such as age-related macular degeneration and macular telangiectasia. Ocular neovascularization is strongly associated with inflammation, but the source of inflammatory signals and the mechanisms by which these signals regulate the disruption of avascular privilege in photoreceptors are unknown. In this study, we found that c-Fos, a master inflammatory regulator, was increased in photoreceptors in a model of pathological blood vessels invading photoreceptors: the very low-density lipoprotein receptor-deficient (Vldlr-/- ) mouse. Increased c-Fos induced inflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor (TNF), leading to activation of signal transducer and activator of transcription 3 (STAT3) and increased TNFα-induced protein 3 (TNFAIP3) in Vldlr-/- photoreceptors. IL-6 activated the STAT3/vascular endothelial growth factor A (VEGFA) pathway directly, and elevated TNFAIP3 suppressed SOCS3 (suppressor of cytokine signaling 3)-activated STAT3/VEGFA indirectly. Inhibition of c-Fos using photoreceptor-specific AAV (adeno-associated virus)-hRK (human rhodopsin kinase)-sh_c-fos or a chemical inhibitor substantially reduced the pathological neovascularization and rescued visual function in Vldlr-/- mice. These findings suggested that the photoreceptor c-Fos controls blood vessel growth into the normally avascular photoreceptor layer through the inflammatory signal-induced STAT3/VEGFA pathway.


Asunto(s)
Inflamación/metabolismo , Inflamación/patología , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Transducción de Señal , Animales , Dependovirus/metabolismo , Interleucina-6/metabolismo , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células Fotorreceptoras de Vertebrados/efectos de los fármacos , Células Fotorreceptoras de Vertebrados/patología , ARN Interferente Pequeño/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo , Retina/efectos de los fármacos , Retina/patología , Retina/fisiopatología , Retinoides/farmacología , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo
13.
Sci Signal ; 8(395): ra94, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26396267

RESUMEN

Neurons and glial cells in the retina contribute to neovascularization, or the formation of abnormal new blood vessels, in proliferative retinopathy, a condition that can lead to vision loss or blindness. We identified a mechanism by which suppressor of cytokine signaling 3 (SOCS3) in neurons and glial cells prevents neovascularization. We found that Socs3 expression was increased in the retinal ganglion cell and inner nuclear layers after oxygen-induced retinopathy. Mice with Socs3 deficiency in neuronal and glial cells had substantially reduced vaso-obliterated retinal areas and increased pathological retinal neovascularization in response to oxygen-induced retinopathy, suggesting that loss of neuronal/glial SOCS3 increased both retinal vascular regrowth and pathological neovascularization. Furthermore, retinal expression of Vegfa (which encodes vascular endothelial growth factor A) was higher in these mice than in Socs3 flox/flox controls, indicating that neuronal and glial SOCS3 suppressed Vegfa expression during pathological conditions. Lack of neuronal and glial SOCS3 resulted in greater phosphorylation and activation of STAT3, which led to increased expression of its gene target Vegfa, and increased endothelial cell proliferation. In summary, SOCS3 in neurons and glial cells inhibited the STAT3-mediated secretion of VEGF from these cells, which suppresses endothelial cell activation, resulting in decreased endothelial cell proliferation and angiogenesis. These results suggest that neuronal and glial cell SOCS3 limits pathological retinal angiogenesis by suppressing VEGF signaling.


Asunto(s)
Neuroglía/metabolismo , Neovascularización Retiniana/metabolismo , Neuronas Retinianas/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Endoteliales/metabolismo , Células Endoteliales/patología , Ratones , Ratones Noqueados , Neuroglía/patología , Oxígeno/toxicidad , Neovascularización Retiniana/inducido químicamente , Neovascularización Retiniana/patología , Neuronas Retinianas/patología , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Factor A de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA