Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 320
Filtrar
1.
Development ; 149(2)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35050341

RESUMEN

Angiogenesis is a stepwise process leading to blood vessel formation. In the vertebrate retina, endothelial cells are guided by astrocytes migrating along the inner surface, and the two processes are coupled by a tightly regulated cross-talks between the two cell types. Here, I have investigated how the FAT1 cadherin, a regulator of tissue morphogenesis that governs tissue cross-talk, influences retinal vascular development. Late-onset Fat1 inactivation in the neural lineage in mice, by interfering with astrocyte progenitor migration polarity and maturation, delayed postnatal retinal angiogenesis, leading to persistent vascular abnormalities in adult retinas. Impaired astrocyte migration and polarity were not associated with alterations of retinal ganglion cell axonal trajectories or of the inner limiting membrane. In contrast, inducible Fat1 ablation in postnatal astrocytes was sufficient to alter their migration polarity and proliferation. Altogether, this study uncovers astrocyte-intrinsic and -extrinsic Fat1 activities that influence astrocyte migration polarity, proliferation and maturation, disruption of which impacts retinal vascular development and maintenance.


Asunto(s)
Astrocitos/metabolismo , Cadherinas/metabolismo , Neovascularización Fisiológica , Retina/metabolismo , Animales , Astrocitos/citología , Cadherinas/genética , Diferenciación Celular , Ratones , Retina/citología , Retina/crecimiento & desarrollo , Vasos Retinianos/crecimiento & desarrollo
2.
Cutan Ocul Toxicol ; 40(4): 359-364, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34429007

RESUMEN

BACKGROUND AND AIM: Retinopathy of prematurity (ROP) is the major ocular problem of preterm infants that occurs with abnormal proliferation of immature retinal vessels. Although pentoxifylline (PTX) was reported to inhibit vasculogenesis and neovascularization in experimental studies, there is no clinical data about the effects of PTX treatment on the development and severity of ROP. This clinical study aimed to investigate the possible effects of PTX on the development of ROP. MATERIALS AND METHODS: A single-centre retrospective study was conducted including preterm infants who were hospitalised in the neonatal intensive care unit between 2015-2017 years. Infants were divided into two groups in terms of PTX administration for adjuvant therapy, as PTX and non-PTX groups. RESULTS: A total of 211 infants were included in the study [gestational age 29 (27-31) weeks, birth weight 1140 (960-1340) g]. From these, 97 infants (46%) were given PTX treatment. The two groups were similar in terms of demographic data and baseline clinical characteristics. Any stage of ROP was detected in 47.4% of infants in the PTX group, which was significantly higher than those in the non-PTX group (27.2%) (p = 0.002). The incidence of advanced-stage ROP in the PTX group (10.3%) was also higher than in the non-PTX group (2.6%) (p = 0.021). Repeated usage of PTX was not found to be related to the development of ROP (p = 0.059). The time of PTX administration was similar between the ROP and no-ROP groups (median; one vs one week, p = 0.825). Surfactant therapy, duration of hospital stay, and PTX treatment were found as significant risk factors for ROP in the logistic regression analysis. CONCLUSIONS: In contrast to the experimental studies and also promising results of PTX treatment in some neonatal morbidities, it may be associated with increased incidence and stage of ROP.


Asunto(s)
Pentoxifilina/administración & dosificación , Vasos Retinianos/efectos de los fármacos , Retinopatía de la Prematuridad/terapia , Terapia Combinada/métodos , Transfusión de Eritrocitos , Femenino , Edad Gestacional , Humanos , Incidencia , Recien Nacido Extremadamente Prematuro , Recién Nacido , Recién Nacido de muy Bajo Peso , Unidades de Cuidado Intensivo Neonatal , Masculino , Oxígeno/administración & dosificación , Surfactantes Pulmonares/administración & dosificación , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/patología , Retinopatía de la Prematuridad/diagnóstico , Retinopatía de la Prematuridad/patología , Estudios Retrospectivos , Factores de Riesgo , Índice de Severidad de la Enfermedad
3.
Dev Biol ; 478: 144-154, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34260962

RESUMEN

Throughout the central nervous system, astrocytes adopt precisely ordered spatial arrangements of their somata and arbors, which facilitate their many important functions. Astrocyte pattern formation is particularly important in the retina, where astrocytes serve as a template that dictates the pattern of developing retinal vasculature. Thus, if astrocyte patterning is disturbed, there are severe consequences for retinal angiogenesis and ultimately for vision - as seen in diseases such as retinopathy of prematurity. Here we discuss key steps in development of the retinal astrocyte population. We describe how fundamental developmental forces - their birth, migration, proliferation, and death - sculpt astrocytes into a template that guides angiogenesis. We further address the radical changes in the cellular and molecular composition of the astrocyte network that occur upon completion of angiogenesis, paving the way for their adult functions in support of retinal ganglion cell axons. Understanding development of retinal astrocytes may elucidate pattern formation mechanisms that are deployed broadly by other axon-associated astrocyte populations.


Asunto(s)
Astrocitos/fisiología , Retina/crecimiento & desarrollo , Retina/fisiología , Animales , Axones/fisiología , Muerte Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Humanos , Neovascularización Fisiológica , Fibras Nerviosas/fisiología , Retina/citología , Retina/embriología , Células Ganglionares de la Retina/fisiología , Vasos Retinianos/embriología , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/fisiología , Retinopatía de la Prematuridad/patología , Retinopatía de la Prematuridad/fisiopatología
4.
J Vis Exp ; (170)2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33871456

RESUMEN

Oxygen-induced retinopathy (OIR) is widely used to study abnormal vessel growth in ischemic retinal diseases, including retinopathy of prematurity (ROP), proliferative diabetic retinopathy (PDR), and retinal vein occlusion (RVO). Most OIR studies observe retinal neovascularization at specific time points; however, the dynamic vessel growth in live mice along a time course, which is essential for understanding the OIR-related vessel diseases, has been understudied. Here, we describe a step-by-step protocol for the induction of the OIR mouse model, highlighting the potential pitfalls, and providing an improved method to quickly quantify areas of vaso-obliteration (VO) and neovascularization (NV) using immunofluorescence staining. More importantly, we monitored vessel regrowth in live mice from P15 to P25 by performing fluorescein fundus angiography (FFA) in the OIR mouse model. The application of FFA to the OIR mouse model allows us to observe the remodeling process during vessel regrowth.


Asunto(s)
Retinopatía Diabética , Modelos Animales de Enfermedad , Neovascularización Retiniana , Vasos Retinianos/crecimiento & desarrollo , Retinopatía de la Prematuridad , Animales , Animales Recién Nacidos , Femenino , Angiografía con Fluoresceína , Masculino , Ratones Endogámicos C57BL , Oxígeno
5.
Biomolecules ; 11(4)2021 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-33919531

RESUMEN

Bone morphogenetic proteins (BMPs) play an important role in bone formation and repair. Recent studies underscored their essential role in the normal development of several organs and vascular homeostasis in health and diseases. Elevated levels of BMPs have been linked to the development of cardiovascular complications of diabetes mellitus. However, their particular role in the pathogenesis of microvascular dysfunction associated with diabetic retinopathy (DR) is still under-investigated. Accumulated evidence from our and others' studies suggests the involvement of BMP signaling in retinal inflammation, hyperpermeability and pathological neovascularization in DR and age-related macular degeneration (AMD). Therefore, targeting BMP signaling in diabetes is proposed as a potential therapeutic strategy to halt the development of microvascular dysfunction in retinal diseases, particularly in DR. The goal of this review article is to discuss the biological functions of BMPs, their underlying mechanisms and their potential role in the pathogenesis of DR in particular.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Retinopatía Diabética/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Humanos , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/metabolismo , Vasos Retinianos/patología , Transducción de Señal
6.
Invest Ophthalmol Vis Sci ; 62(2): 21, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33595613

RESUMEN

Purpose: Pathological blood vessel growth in the eye is implicated in several diseases that result in vision loss, including age-related macular degeneration and diabetic retinopathy. The limits of current disease therapies have created the need to identify and characterize new antiangiogenic drugs. Here, we identify the secreted chemorepellent semaphorin-3fa (Sema3fa) as an endogenous anti-angiogenic in the eye. Methods: We generated a CRISPR/Cas9 sema3fa zebrafish mutant line, sema3faca304/304. We assessed the retinal and choroidal vasculature in both larval and adult wild-type and sema3fa mutant zebrafish. Results: We find sema3fa mRNA is expressed by the ciliary marginal zone, neural retina, and retinal pigment epithelium of zebrafish larvae as choroidal vascularization emerges and the hyaloid/retinal vasculature is remodeled. The hyaloid vessels of sema3fa mutants develop appropriately but fail to remodel during the larval period, with adult mutants exhibiting a denser network of capillaries in the retinal periphery than seen in wild-type. The choroid vasculature is also defective in that it develops precociously, and aberrant, leaky sprouts are present in the normally avascular outer retina of both sema3faca304/304 larvae and adult fish. Conclusions: Sema3fa is a key endogenous signal for maintaining an avascular retina and preventing pathologic vascularization. Furthermore, we provide a new experimentally accessible model for studying choroid neovascularization (CNV) resulting from primary changes in the retinal environment that lead to downstream vessel infiltration.


Asunto(s)
Capilares/crecimiento & desarrollo , ADN/genética , Degeneración Macular/genética , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Epitelio Pigmentado de la Retina/metabolismo , Vasos Retinianos/crecimiento & desarrollo , Animales , Capilares/metabolismo , Coroides/metabolismo , Coroides/patología , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Degeneración Macular/metabolismo , Degeneración Macular/patología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Epitelio Pigmentado de la Retina/patología , Vasos Retinianos/metabolismo , Pez Cebra
7.
Angiogenesis ; 24(2): 363-377, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33201372

RESUMEN

NADPH oxidase 4 (Nox4) is a major isoform of NADPH oxidases playing an important role in many biological processes. Previously we have shown that Nox4 is highly expressed in retinal blood vessels and is upregulated in oxygen-induced retinopathy (OIR). However, the exact role of endothelial Nox4 in retinal angiogenesis remains elusive. Herein, using endothelial cell (EC)-specific Nox4 knockout (Nox4EC-KO) mice, we investigated the impact of endothelial Nox4 deletion on retinal vascular development and pathological angiogenesis during OIR. Our results show that deletion of Nox4 in ECs led to retarded retinal vasculature development with fewer, blunted-end tip cells and sparser, dysmorphic filopodia at vascular front, and reduced density of vascular network in superficial, deep, and intermediate layers in postnatal day 7 (P7), P12, and P17 retinas, respectively. In OIR, loss of endothelial Nox4 had no effect on hyperoxia-induced retinal vaso-obliteration at P9 but significantly reduced aberrant retinal neovascularization at P17 and decreased the deep layer capillary density at P25. Ex vivo study confirmed that lack of Nox4 in ECs impaired vascular sprouting. Mechanistically, loss of Nox4 significantly reduced expression of VEGF, p-VEGFR2, integrin αV, angiopoietin-2, and p-ERK1/2, attenuating EC migration and proliferation. Taken together, our results indicate that endothelial Nox4 is important for retinal vascular development and contributes to pathological angiogenesis, likely through regulation of VEGF/VEGFR2 and angiopoietin-2/integrin αV/ERK pathways. In addition, our study suggests that endothelial Nox4 appears to be essential for intraretinal revascularization after hypoxia. These findings call for caution on targeting endothelial Nox4 in ischemic/hypoxic retinal diseases.


Asunto(s)
Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Eliminación de Gen , NADPH Oxidasa 4/metabolismo , Neovascularización Fisiológica , Neovascularización Retiniana/enzimología , Vasos Retinianos/crecimiento & desarrollo , Animales , Ratones , Ratones Noqueados , NADPH Oxidasa 4/genética , Neovascularización Retiniana/genética
8.
PLoS One ; 15(12): e0242667, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33259500

RESUMEN

BACKGROUND: To prospectively investigate retinal vascular changes in patients undergoing epiretinal membrane (ERM) and internal limiting membrane (ILM) peeling using swept source optical coherence tomography angiography (SSOCTA). METHODS: Consecutive patients were grouped based on ERM severity and followed using SSOCTA up to month 3 after surgical intervention. Superficial and deep foveal avascular zone (s/dFAZ) as well as foveal and parafoveal vessel density (VD) were correlated with ERM severity and visual acuity. Differences between groups were evaluated. RESULTS: Significant correlations were found between ERM severity and baseline sFAZ, dFAZ and best corrected visual acuity (BCVA), central retinal subfield thickness (CST) and ΔCST (r = -0.52, r = -0.43, r = -0.42, r = 0.58, r = 0.39; all p<0.05). Vascular flow parameters did not correlate with age, peeling size, pseudophakia or CST, but correlated with intraretinal cysts presence. No associations of BCVA with any of the OCTA parameters across time were found. Significant differences between ERM severity groups 1 and 2 were found for sFAZ at baseline (p = 0.005) and at the 3-month follow-up (p = 0.014), and for dFAZ at baseline (p = 0.017). Superficial foveal and parafoveal VD were not significantly different between groups (all p>0.05). CONCLUSIONS: This study clearly shows that ERM severity based on ERM staging has to be taken into account when undertaking studies in patients with idiopathic ERM using SSOCTA. Further, specific changes in the superficial and deep retinal vasculature in eyes undergoing ERM and ILM peeling were found. However, the clinical usefulness and prognostic value for post-surgical treatment BCVA of the SSOCTA-derived variables (sFAZ and dFAZ area, as well as foveal and parafoveal VD) used remains questionable.


Asunto(s)
Membrana Epirretinal/fisiopatología , Retina/diagnóstico por imagen , Vasos Retinianos/diagnóstico por imagen , Tomografía de Coherencia Óptica , Anciano , Vasos Sanguíneos/diagnóstico por imagen , Vasos Sanguíneos/crecimiento & desarrollo , Vasos Sanguíneos/fisiopatología , Membrana Epirretinal/diagnóstico por imagen , Femenino , Angiografía con Fluoresceína , Fóvea Central/irrigación sanguínea , Fóvea Central/fisiopatología , Humanos , Mácula Lútea/irrigación sanguínea , Mácula Lútea/diagnóstico por imagen , Mácula Lútea/fisiopatología , Masculino , Persona de Mediana Edad , Retina/fisiopatología , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/fisiopatología , Agudeza Visual/fisiología , Vitrectomía
9.
Sci Rep ; 10(1): 18601, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139746

RESUMEN

To investigate the effects of a high-fat diet (HFD) and apolipoprotein E (Apoe) deficiency on retinal structure and function in mice. Apoe KO mice and wild-type C57BL/6J mice were given a low-fat diet (LFD) or a HFD for 32 weeks. Blood glucose, serum lipids, body weight and visceral fat weight were evaluated. Retinal sterol quantification was carried out by isotope dilution gas chromatography-mass spectrometry. The cholesterol metabolism related genes SCAP-SREBP expressions were detected by qRT-PCR. Retinal function was recorded using an electroretinogram. The thickness of each layer of the retina was measured by optical coherence tomography. Fundus fluorescein angiography was performed to detect retinal vasculature changes. Immunohistochemical staining was used to determine the expression of NF-κB, TNF-α and VEGFR2 in the retina among HFD, HFD Apoe-/-, LFD Apoe-/- and WT mice retinas. HFD feeding caused the mice to gain weight and develop hypercholesterinemia, while Apoe-/- abnormalities also affected blood lipid metabolism. Both HFD and Apoe deficiency elevated retinal cholesterol, especially in the HFD Apoe-/- mice. No up-regulated expression of SCAP-SREBP was observed as a negative regulator. Impaired retinal functions, thinning retinas and abnormal retinal vasculature were observed in the peripheral retinas of the HFD and Apoe-/- mice compared with those in the normal chow group, particularly in the HFD Apoe-/- mice. Moreover, the expression of NF-κB in the retinas of the HFD and Apoe-/- mice was increased, together with upregulated TNF-α mRNA levels and TNF-α expression in the layer of retinal ganglion cells of the peripheral retina. At the same time, the expression level of VEGFR2 was elevated in the intervention groups, most notably in HFD Apoe-/- mice. HFD or Apoe gene deletion had certain adverse effects on retinal function and structure, which were far below the combined factors and induced harm to the retina. Furthermore, HFD caused retinal ischemia and hypoxia. Additionally, Apoe abnormality increased susceptibility to ischemia. These changes upregulated NF-κB expression in ganglion cells and activated downstream TNF-α. Simultaneously, they activated VEGFR2, accelerating angiogenesis and vascular permeability. All of the aforementioned outcomes initiated inflammatory responses to trigger ganglion cell apoptosis and aggravate retinal neovascularization.


Asunto(s)
Apolipoproteínas E/genética , Dieta Alta en Grasa , Retina/patología , Retina/fisiología , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica , Vasos Retinianos/crecimiento & desarrollo
10.
Sci Rep ; 10(1): 12901, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32733052

RESUMEN

This study investigates the hyaloid vascular regression and its relationship to the retinal and choroidal vascular developments using optical coherence tomography angiography (OCTA). Normal and oxygen-induced retinopathy (OIR) rat eyes at postnatal day 15, 18, 21, and 24 were longitudinally imaged using OCTA. At each day, two consecutive imaging for visualizing the hyaloid vasculature and the retinal and choroidal vasculatures were conducted. The hyaloid vessel volume and the retinal and choroidal vessel densities were measured. The hyaloid vessel volumes gradually decreased during the regression, although the OIR eyes exhibited large vessel volumes at all time points. A spatial relationship between persistent hyaloid vasculature and retardation of underlying retinal vascular development was observed in the OIR eyes. Furthermore, anti-vascular endothelial growth factor (VEGF) was administered intravitreally to additional OIR eyes to observe its effect on the vascular regression and development. The VEGF injection to OIR eyes showed reduced persistent hyaloid vessels in the injected eyes as well as in the non-injected fellow eyes. This study presents longitudinal imaging of intraocular vasculatures in the developing eye and shows the utility of OCTA that can be widely used in studies of vascular development and regression and preclinical evaluation of new anti-angiogenic drugs.


Asunto(s)
Coroides , Angiografía con Fluoresceína , Neovascularización Fisiológica , Vasos Retinianos , Tomografía de Coherencia Óptica , Animales , Coroides/irrigación sanguínea , Coroides/diagnóstico por imagen , Femenino , Masculino , Ratas , Ratas Sprague-Dawley , Vasos Retinianos/diagnóstico por imagen , Vasos Retinianos/crecimiento & desarrollo , Factor A de Crecimiento Endotelial Vascular/farmacología
11.
CRISPR J ; 3(4): 284-298, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32833527

RESUMEN

Humans carrying homozygous loss-of-function mutations in the Wnt co-receptor, low-density lipoprotein receptor-related protein 5 (LRP5), develop osteoporosis and a defective retinal vasculature known as familial exudative vitreoretinopathy (FEVR) due to disruption of the Wnt signaling pathway. The purpose of this study was to use CRISPR-Cas9-mediated gene editing to create strains of Lrp5-deficient rats and to determine whether knockout of Lrp5 resulted in phenotypes that model the bone and retina pathology in LRP5-deficient humans. Knockout of Lrp5 in rats produced low bone mass, decreased bone mineral density, and decreased bone size. The superficial retinal vasculature of Lrp5-deficient rats was sparse and disorganized, with extensive exudates and decreases in vascularized area, vessel length, and branch point density. This study showed that Lrp5 could be predictably knocked out in rats using CRISPR-Cas9, causing the expression of bone and retinal phenotypes that will be useful for studying the role of Wnt signaling in bone and retina development and for research on the treatment of osteoporosis and FEVR.


Asunto(s)
Huesos/metabolismo , Técnicas de Inactivación de Genes , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Vasos Retinianos/metabolismo , Animales , Huesos/fisiopatología , Femenino , Regulación de la Expresión Génica , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Mutación , Ratas , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/fisiopatología , Vía de Señalización Wnt
12.
Sci Rep ; 10(1): 9299, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32518264

RESUMEN

Ischemic proliferative retinopathy (IPR), such as proliferative diabetic retinopathy (PDR), retinal vein occlusion and retinopathy of prematurity is a major cause of vision loss. Our previous studies demonstrated that periostin (PN) and tenascin-C (TNC) are involved in the pathogenesis of IPR. However, the interactive role of PN and TNC in angiogenesis associated with IPR remain unknown. We found significant correlation between concentrations of PN and TNC in PDR vitreous humor. mRNA and protein expression of PN and TNC were found in pre-retinal fibrovascular membranes excised from PDR patients. Interleukin-13 (IL-13) promoted mRNA and protein expression of PN and TNC, and co-immunoprecipitation assay revealed binding between PN and TNC in human microvascular endothelial cells (HRECs). IL-13 promoted angiogenic functions of HRECs. Single inhibition of PN or TNC and their dual inhibition by siRNA suppressed the up-regulated angiogenic functions. Pathological pre-retinal neovessels of oxygen-induced retinopathy (OIR) mice were attenuated in PN knock-out, TNC knock-out and dual knock-out mice compared to wild-type mice. Both in vitro and in vivo, PN inhibition had a stronger inhibitory effect on angiogenesis compared to TNC inhibition, and had a similar effect to dual inhibition of PN and TNC. Furthermore, PN knock-out mice showed scant TNC expression in pre-retinal neovessels of OIR retinas. Our findings suggest that interaction of PN and TNC facilitates pre-retinal angiogenesis, and PN is an effective therapeutic target for IPR such as PDR.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Retinopatía Diabética/patología , Neovascularización Patológica/patología , Vasos Retinianos/crecimiento & desarrollo , Tenascina/metabolismo , Vitreorretinopatía Proliferativa/patología , Anciano , Animales , Moléculas de Adhesión Celular/genética , Células Cultivadas , Células Endoteliales/metabolismo , Femenino , Humanos , Interleucina-13/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Tenascina/genética , Cuerpo Vítreo/metabolismo
13.
Biol Pharm Bull ; 43(5): 859-863, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32378561

RESUMEN

Pathological angiogenesis is a leading cause of blindness in several retinal diseases. The key driving factor inducing pathological angiogenesis is the pronounced hypoxia leading to a marked, increased production of vascular endothelial growth factor (VEGF). The aim of this study was to determine whether the abnormal vascular growth occurs in a manner dependent on the degree of the vascular defects. Vascular defects of two different degrees were created in the retina by subcutaneously treating neonatal rats with the VEGF receptor (VEGFR) tyrosine kinase inhibitor KRN633 on postnatal day (P) 4 and P5 (P4/5) or P7 and P8 (P7/8). The structure of the retinal vasculature changes was examined immunohistochemically. Prevention of vascular growth and regression of some preformed capillaries were observed on the next day, after completion of each treatment (i.e., P6 and P9). The vascular regrowth occurred as a result of eliminating the inhibitory effect on the VEGFR signaling pathway. KRN633 (P4/5)-treated rats exhibited a retinal vasculature with aggressive intravitreal neovascularization on P21. On the other hand, the appearance of tortuous arteries is a representative vascular pathological feature in retinas of KRN633 (P7/8)-treated groups. These results suggest that an interruption of the retinal vascular development at different time points induces different vascular pathological features in the retina. Pharmacological agents targeting the VEGF signaling pathway are useful for creating an abnormal retinal vasculature with various pathological features in order to evaluate the efficacy of anti-angiogenic compounds.


Asunto(s)
Compuestos de Fenilurea/administración & dosificación , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinazolinas/administración & dosificación , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Vasos Retinianos/efectos de los fármacos , Animales , Animales Recién Nacidos , Fenotipo , Ratas Sprague-Dawley , Vasos Retinianos/crecimiento & desarrollo , Vasos Retinianos/patología , Factores de Tiempo
14.
Am J Ophthalmol ; 219: 231-239, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32413409

RESUMEN

PURPOSE: To quantify the macular microvasculature in healthy children of various ages by using optical coherence tomography angiography (OCTA). DESIGN: Prospective cross-sectional study. METHODS: A total of 333 normal children from 4 to 16 years old were included. OCTA was performed on a 3- × 3-mm area centered on the macular region. Vascular density, perfusion density, fovea avascular zone (FAZ) area, FAZ perimeter, and FAZ acircularity index (AI) were measured and adjusted for axial length. Differences were compared among various ages. RESULTS: Among the different age groups, both macular vascular density and perfusion density increased with age (P < .0001 and P = .0028, respectively). After adjustments were made for the spherical equivalent (SE) and axial length, macular vascular density was significantly associated with age (r = 0.183; P = .001) No factors were significantly correlated with the perfusion density after adjustment for the age, SE, or axial length. The FAZ area and FAZ perimeter did not change among groups of different ages. Nevertheless, the AI of FAZ in the 4.00-6.99-year-old group was smaller to that of the 13.00-15.99-year-old group (P = .03). Younger children had significantly higher rates of nonconsecutive vessels branched toward the macular center (P = .0002) and vascular loops contributing to irregular shapes of FAZ (P = .024). CONCLUSIONS: Macular vascular density and perfusion density continuously increase with age in children. Despite the fact that FAZ area and perimeter did not change, the microstructure of FAZ pruned and tended to form a smooth and regular avascular area during development.


Asunto(s)
Angiografía con Fluoresceína , Microvasos/crecimiento & desarrollo , Vasos Retinianos/crecimiento & desarrollo , Tomografía de Coherencia Óptica , Adolescente , Niño , Preescolar , Estudios Transversales , Femenino , Fóvea Central/irrigación sanguínea , Voluntarios Sanos , Humanos , Masculino , Microvasos/diagnóstico por imagen , Estudios Prospectivos , Valores de Referencia , Vasos Retinianos/diagnóstico por imagen , Agudeza Visual/fisiología
15.
J Neuroendocrinol ; 32(11): e12858, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32449569

RESUMEN

The hormone prolactin (PRL) is emerging as an important regulator of ocular blood vessels. PRL is pro-angiogenic and acquires anti-angiogenic properties after undergoing proteolytic cleavage to the PRL fragment, vasoinhibin. The vascularisation of the rodent retina develops after birth when it rapidly expands until completion at the end of the first postnatal week. Exposure of newborn mice to high oxygen levels lowers the rate of blood vessel growth. In the present study, we investigated whether PRL treatment modifies the vascularisation of the retina in newborn mice exposed to high oxygen or to normoxia and whether the retinal conversion of PRL to vasoinhibin may be altered in the neonate. Newborn mice and their nursing mothers were subjected to 75% oxygen or to normoxia from postnatal day (P) 6 to P8 (group 1) or from P2 to P5 (group 2). PRL (2 µg g-1 , i.p., twice a day) or vehicle was injected from P5 to P8 in group 1 and from P1 to P5 in group 2. PRL treatment reduced the retinal inhibition of blood vessel growth and the increase in vascular regression induced by hyperoxia as revealed by immunofluorescence staining of blood vessels and the expression of angiogenesis and apoptosis markers. The pro-angiogenic effect may involve a reduced conversion of PRL to vasoinhibin. Incubation of PRL with retinal extracts showed reduced activity of the PRL-cleaving protease, cathepsin D, in the neonate vs the adult retina that was further reduced under hyperoxia. PRL and the PRL receptor mRNA were expressed at higher levels in the retina at P8 than in the adult, whereas endogenous PRL was undetectable in the circulation at P8. We conclude that PRL has a pro-angiogenic effect in the neonate retina as a result of its reduced conversion to vasoinhibin and that PRL produced by the retina may help promote physiological vascularisation after birth.


Asunto(s)
Hiperoxia , Neovascularización Fisiológica , Prolactina , Vasos Retinianos , Animales , Femenino , Masculino , Ratones , Embarazo , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Hiperoxia/patología , Neovascularización Fisiológica/efectos de los fármacos , Prolactina/sangre , Prolactina/metabolismo , Prolactina/farmacología , Receptores de Prolactina/efectos de los fármacos , Receptores de Prolactina/metabolismo , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/crecimiento & desarrollo , Retinopatía de la Prematuridad/patología
16.
Nat Commun ; 11(1): 986, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32080187

RESUMEN

Tissue microarchitecture and mechanics are important in development and pathologies of the Central Nervous System (CNS); however, their coordinating mechanisms are unclear. Here, we report that during colonization of the retina, microglia contacts the deep layer of high stiffness, which coincides with microglial bipolarization, reduction in TGFß1 signaling and termination of vascular growth. Likewise, stiff substrates induce microglial bipolarization and diminish TGFß1 expression in hydrogels. Both microglial bipolarization in vivo and the responses to stiff substrates in vitro require intracellular adaptor Kindlin3 but not microglial integrins. Lack of Kindlin3 causes high microglial contractility, dysregulation of ERK signaling, excessive TGFß1 expression and abnormally-patterned vasculature with severe malformations in the area of photoreceptors. Both excessive TGFß1 signaling and vascular defects caused by Kindlin3-deficient microglia are rescued by either microglial depletion or microglial knockout of TGFß1 in vivo. This mechanism underlies an interplay between microglia, vascular patterning and tissue mechanics within the CNS.


Asunto(s)
Microglía/fisiología , Vasos Retinianos/inervación , Factor de Crecimiento Transformador beta1/fisiología , Actomiosina/fisiología , Animales , Fenómenos Biomecánicos , Movimiento Celular/fisiología , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/fisiología , Femenino , Hidrogeles , Integrinas/fisiología , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microglía/citología , Comunicación Paracrina , Retina/crecimiento & desarrollo , Vasos Retinianos/citología , Vasos Retinianos/crecimiento & desarrollo , Factor de Crecimiento Transformador beta1/genética
17.
Biochem Biophys Res Commun ; 521(4): 939-946, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31718797

RESUMEN

Crumbs proteins are transmembrane proteins that regulate cellular apico-basal polarity. Animals carrying mutated crb1 present retinal vascular abnormalities; this mutation is associated with progressive retinal degeneration with intraretinal cystoid fluid collection in humans. This study aimed to evaluate a potential role of crumbs proteins in retinal vascular development and maintenance. We demonstrated that crumbs homologues (CRBs) were differentially expressed and changed dramatically during mouse retinal vascular development. Intravitreal injection of CRB1 and CRB2 siRNA induced delayed development of the deep capillary plexus and premature development of the intermediate capillary plexus, resulting in disrupted vascular integrity. However, microfluidic chip assay using human retinal endothelial cells revealed that CRBs do not directly affect in vitro retinal angiogenesis. CRBs control retinal angiogenesis by regulating neuroglial vascular endothelial growth factor-A (VEGFA) and matrix metalloproteinase-3 expression. These findings demonstrate a pivotal role of CRBs in providing critical neurotrophic support through normal layered vascular network development and maintenance. This implies that preserving CRBs and restoring layered retinal vascular networks could be novel targets for preventing vision-threatening retinal diseases.


Asunto(s)
Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Vasos Retinianos/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Células Cultivadas , Electrorretinografía , Células Endoteliales , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Metaloproteinasa 3 de la Matriz/metabolismo , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Mutantes , Neovascularización Fisiológica/genética , Proteínas del Tejido Nervioso/genética , Retina/citología , Retina/efectos de los fármacos , Retina/patología , Vasos Retinianos/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología
18.
Biomed Res Int ; 2019: 6512195, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31828115

RESUMEN

PURPOSE: To investigate the longitudinal findings of fundus features and spectral-domain optical coherence tomography (SD-OCT) to characterize the morphologic features in a mouse model of defective glutamate/aspartate transporter (GLAST-/- mice). MATERIALS AND METHODS: The fundus findings and SD-OCT images were longitudinally recorded at five time points from postnatal (P) 22 to P156 in GLAST-/- mice. As a control wild type, age-matched C57BL/6J mice were employed. The mouse retina was subdivided into five layers, and the thickness of each layer was longitudinally measured by InSight® using SD-OCT pictures. The SD-OCT findings were compared with the histologic appearances. The diameter of the retinal blood vessels was measured by the ImageJ® software program using SD-OCT images. The data were statistically compared between both age-matched mouse groups. RESULTS: The retinal blood vessels appeared more dilated in GLAST-/- mice than in wild-type mice. This tendency was statistically significant at all time points after P44 by analyses using SD-OCT images. The ganglion cell complex (GCC) and outer nuclear layer (ONL) were significantly thinner in GLAST-/- mice at all time points after P80 than in the wild-type mice. This tendency was more clearly indicated by SD-OCT than histologic sections. DISCUSSION: In the present study, we found for the first time the dilation of the retinal blood vessels and the thinning of the ONL in GLAST-/- mice, in addition to the thinning of the GCC.


Asunto(s)
Sistema de Transporte de Aminoácidos X-AG/genética , Ácido Glutámico/metabolismo , Retina/metabolismo , Vasos Retinianos/metabolismo , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Fondo de Ojo , Humanos , Ratones , Ratones Noqueados , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Retina/diagnóstico por imagen , Retina/crecimiento & desarrollo , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Vasos Retinianos/diagnóstico por imagen , Vasos Retinianos/crecimiento & desarrollo , Tomografía de Coherencia Óptica
19.
Nat Commun ; 10(1): 5243, 2019 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-31748531

RESUMEN

Familial exudative vitreoretinopathy (FEVR) is a human disease characterized by defective retinal angiogenesis and associated complications that can result in vision loss. Defective Wnt/ß-catenin signaling is an established cause of FEVR, whereas other molecular alterations contributing to the disease remain insufficiently understood. Here, we show that integrin-linked kinase (ILK), a mediator of cell-matrix interactions, is indispensable for retinal angiogenesis. Inactivation of the murine Ilk gene in postnatal endothelial cells results in sprouting defects, reduced endothelial proliferation and disruption of the blood-retina barrier, resembling phenotypes seen in established mouse models of FEVR. Retinal vascularization defects are phenocopied by inducible inactivation of the gene for α-parvin (Parva), an interactor of ILK. Screening genomic DNA samples from exudative vitreoretinopathy patients identifies three distinct mutations in human ILK, which compromise the function of the gene product in vitro. Together, our data suggest that defective cell-matrix interactions are linked to Wnt signaling and FEVR.


Asunto(s)
Barrera Hematorretinal/metabolismo , Células Endoteliales/metabolismo , Vitreorretinopatías Exudativas Familiares/genética , Neovascularización Fisiológica/genética , Proteínas Serina-Treonina Quinasas/genética , Vasos Retinianos/crecimiento & desarrollo , Animales , Células Endoteliales/citología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Proteínas de Microfilamentos/genética , Fenotipo , Vía de Señalización Wnt/genética
20.
J Clin Invest ; 129(9): 3807-3820, 2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31403471

RESUMEN

Vascular development in the mammalian retina is a paradigm for CNS vascular development in general, and its study is revealing fundamental mechanisms that explain the efficacy of antiangiogenic therapies in retinal vascular disease. During development of the mammalian retina, hypoxic astrocytes are hypothesized to secrete VEGF, which attracts growing endothelial cells as they migrate radially from the optic disc. However, published tests of this model using astrocyte-specific deletion of Vegf in the developing mouse retina appear to contradict this theory. Here, we report that selectively eliminating Vegf in neonatal retinal astrocytes with a Gfap-Cre line that recombines with approximately 100% efficiency had no effect on proliferation or radial migration of astrocytes, but completely blocked radial migration of endothelial cells, strongly supporting the hypoxic astrocyte model. Using additional Cre driver lines, we found evidence for essential and partially redundant actions of retina-derived (paracrine) and astrocyte-derived (autocrine) VEGF in controlling astrocyte proliferation and migration. We also extended previous studies by showing that HIF-1α in retinal neurons and HIF-2α in Müller glia play distinct roles in retinal vascular development and disease, adding to a growing body of data that point to the specialization of these 2 hypoxia-sensing transcription factors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neovascularización Fisiológica , Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Astrocitos/metabolismo , Encéfalo/irrigación sanguínea , Movimiento Celular , Proliferación Celular , Corteza Cerebral/metabolismo , Células Endoteliales/metabolismo , Genotipo , Proteínas Fluorescentes Verdes/metabolismo , Hipoxia , Ratones , Fenotipo , Vasos Retinianos/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA