Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Eur J Pharmacol ; 970: 176510, 2024 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-38493917

RESUMEN

Activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway protects against N-methyl-D-aspartic acid (NMDA)-induced excitotoxic retinal injury. AMPK activation enhances fatty acid metabolism and ketone body synthesis. Ketone bodies are transported into neurons by monocarboxylate transporters (MCTs) and exert neuroprotective effects. In this study, we examined the distribution and expression levels of MCT1 and MCT2 in the retina and analyzed the effects of pharmacological inhibition of MCTs on the protective effects of metformin and 5-aminoimidazole-4-carboxamide (AICAR), activators of AMPK, against NMDA-induced retinal injury in rats. MCT1 was expressed in the blood vessels, processes of astrocytes and Müller cells, and inner segments of photoreceptors in the rat retina, whereas MCT2 was expressed in neuronal cells in the ganglion cell layer (GCL) and in astrocyte processes. The expression levels of MCT2, but not MCT1, decreased one day after intravitreal injection of NMDA (200 nmol). Intravitreal injection of NMDA decreased the number of cells in the GCL compared to the vehicle seven days after injection. Simultaneous injection of metformin (20 nmol) or AICAR (50 nmol) with NMDA attenuated NMDA-induced cell loss in the GCL, and these protective effects were attenuated by AR-C155858 (1 pmol), an inhibitor of MCTs. AR-C155858 alone had no significant effect on the retinal structure. These results suggest that AMPK-activating compounds protect against NMDA-induced excitotoxic retinal injury via mechanisms involving MCTs in rats. NMDA-induced neurotoxicity may be associated with retinal neurodegenerative changes in glaucoma and diabetic retinopathy. Therefore, AMPK-activating compounds may be effective in managing these retinal diseases.


Asunto(s)
Metformina , Enfermedades de la Retina , Tiofenos , Uracilo/análogos & derivados , Ratas , Animales , Proteínas Quinasas Activadas por AMP/metabolismo , N-Metilaspartato/toxicidad , Ratas Sprague-Dawley , Retina/metabolismo , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/prevención & control , Enfermedades de la Retina/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Metformina/efectos adversos
2.
Curr Eye Res ; 49(5): 505-512, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38251680

RESUMEN

PURPOSE: Metformin, a biguanide antihyperglycemic drug, can exert various beneficial effects in addition to its glucose-lowering effect. The effects of metformin are mainly mediated by AMP-activated protein kinase (AMPK)-dependent pathway. AMPK activation interferes with the action of the mammalian target of rapamycin complex 1 (mTORC1), and blockade of mTORC1 pathway suppresses pathological retinal angiogenesis. Therefore, in this study, we examined the effects of metformin on pathological angiogenesis and mTORC1 activity in the retinas of mice with oxygen-induced retinopathy (OIR). METHODS: OIR was induced by exposing the mice to 80% oxygen from postnatal day (P) 7 to P10. The OIR mice were treated with metformin, rapamycin (an inhibitor of mTORC1), or the vehicle from P10 to P12 or P14. The formation of neovascular tufts, revascularization in the central avascular areas, expression of vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) 2, and phosphorylated ribosomal protein S6 (pS6), a downstream indicator of mTORC1 activity, were evaluated at P10, P13, or P15. RESULTS: Neovascular tufts and vascular growth in the central avascular areas were observed in the retinas of P15 OIR mice. The formation of neovascular tufts, but not the revascularization in the central avascular areas, was attenuated by metformin administration from P10 to P14. Metformin had no significant inhibitory effect on the expression of VEGF and VEGFR2, but it reduced the pS6 immunoreactivity in vascular cells at the sites of angiogenesis. Rapamycin completely blocked the phosphorylation of ribosomal protein S6 and markedly reduced the formation of neovascular tufts. CONCLUSIONS: These results suggest that metformin partially suppresses the formation of neovascular tufts on the retinal surface by blocking the mTORC1 signaling pathway. Metformin may exert beneficial effects against the progression of ocular diseases in which abnormal angiogenesis is associated with the pathogenesis.


Asunto(s)
Metformina , Enfermedades de la Retina , Neovascularización Retiniana , Animales , Ratones , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína S6 Ribosómica , Metformina/efectos adversos , Proteínas Quinasas Activadas por AMP/metabolismo , Angiogénesis , Neovascularización Patológica , Enfermedades de la Retina/complicaciones , Transducción de Señal , Oxígeno , Sirolimus/farmacología , Sirolimus/uso terapéutico , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/prevención & control , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Mamíferos/metabolismo
3.
Microvasc Res ; 152: 104626, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-37963514

RESUMEN

Retinopathy of prematurity (ROP), a retinal disease that can occur in premature infants, can lead to severe visual impairment. In this study, we examined the preventive and therapeutic effects of mammalian target of rapamycin complex 1 (mTORC1) inhibition on abnormal retinal blood vessels in a rat model of ROP. To induce ROP-like vascular abnormalities, rats were subcutaneously treated with KRN633, an inhibitor of vascular endothelial growth factor (VEGF) receptor tyrosine kinase, on postnatal day 7 (P7) and P8. KRN633-treated (ROP) rats were treated subcutaneously with the mTORC1 inhibitor rapamycin according to preventive and therapeutic protocols, i.e., from P11 to P13 (P11-P13) and from P14 to P20 (P14-P20), respectively. To compare with the effects of VEGF inhibition, KRN633 was administered according to similar protocols. Changes in retinal vasculature, phosphorylated ribosomal protein S6 (pS6), a downstream indicator of mTORC1 activity, and the proliferative status of vascular cells were evaluated at P14 and P21 using immunohistochemistry. Rapamycin treatment from P11 to P13 prevented increases in arteriolar tortuosity, capillary density, and the number of proliferating vascular cells, and eliminated pS6 immunoreactivity in ROP rats. KRN633 treatment at P11 and P12 (P11/P12) also prevented the appearance of ROP-like retinal blood vessels. Rapamycin treatment from P14 to P20 failed to attenuate arteriolar tortuosity but prevented increases in capillary density and proliferating vascular cell number at the vascular front, but not at the central zone. KRN633 treatment from P14 to P20 significantly reduced abnormalities in the retinal vasculature; however, the effects were inferior to those of KRN633 treatment on P11/P12. These results suggest that activation of the mTORC1 pathway in proliferating endothelial cells contributes to the appearance and progression of ROP-like retinal blood vessels. Therefore, inhibition of mTORC1 may be a promising approach for selectively targeting abnormal retinal blood vessels in ROP.


Asunto(s)
Compuestos de Fenilurea , Quinazolinas , Retinopatía de la Prematuridad , Animales , Ratas , Animales Recién Nacidos , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/farmacología , Vasos Retinianos , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/prevención & control , Sirolimus/farmacología , Sirolimus/metabolismo , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Microvasc Res ; 146: 104455, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36396077

RESUMEN

Methylglyoxal, a highly reactive dicarbonyl compound, is increased and accumulated in patients with diabetic mellitus. Methylglyoxal forms advanced glycation end products (AGE), contributing to the pathogenesis of diabetic complications, including diabetic retinopathy. Recent studies have shown that methylglyoxal induces diabetic retinopathy-like abnormalities in retinal vasculature. In this study, we investigated the processes and mechanisms of methylglyoxal-induced retinal capillary endothelial cell degeneration in rats. Morphological changes in vascular components (endothelial cells, pericytes, and basement membranes) were assessed in the retinas 2, 7, and 14 days after intravitreal injection of methylglyoxal. Intravitreal methylglyoxal injection induced retinal capillary endothelial cell degeneration in a dose- and time-dependent manner. Changes in the shape and distribution of pericytes occurred before the initiation of capillary regression in the retinas of methylglyoxal-injected eyes. The receptor for AGEs (RAGEs) antagonist FPS-ZM1, and the matrix metalloproteinase (MMP) inhibitor GM6001 significantly attenuated methylglyoxal-induced capillary endothelial cell degeneration. FPS-ZM1 failed to prevent pathological changes in pericytes in methylglyoxal-injected eyes. In situ zymography revealed that MMP activity was enhanced at sites of blood vessels with reduced pericyte coverage in methylglyoxal-injected eyes. These results suggest that intravitreal methylglyoxal injection induces pathological changes in pericytes before the initiation of capillary endothelial cell degeneration via an AGE-RAGE-independent pathway. The capillary endothelial cell degeneration is mediated by activating the AGE-RAGE pathway and increasing MMP activity in endothelial cells by impairing pericyte function in the retina.


Asunto(s)
Retinopatía Diabética , Ratas , Animales , Retinopatía Diabética/metabolismo , Piruvaldehído/toxicidad , Piruvaldehído/metabolismo , Células Endoteliales/metabolismo , Retina/metabolismo , Vasos Retinianos/patología , Pericitos/metabolismo
5.
Neurosci Lett ; 793: 136999, 2023 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-36470506

RESUMEN

Resveratrol, a natural polyphenolic compound, reportedly possesses numerous biological activities, including anti-inflammatory and antioxidant effects. In the current study, we examined (1) the dilator effects of resveratrol on retinal arterioles, (2) the protective effects of resveratrol against excitotoxic retinal injury, and (3) whether these effects are mediated by the AMP-activated kinase (AMPK)-dependent pathway in rats. Male Wistar rats (7 to 10 weeks old) were used in this study. The diameters of the retinal arterioles, mean arterial pressure, and heart rate were measured in vivo. The retinal injury was assessed by histological examination. Intravenous injection of resveratrol (3 mg/kg) increased the diameter of the retinal arterioles without affecting the mean arterial pressure and heart rate. The AMPK inhibitor, compound C (5 mg/kg, intravenously), significantly attenuated the retinal vasodilator response to resveratrol. Seven days after intravitreal injection of N-methyl-d-aspartic acid (NMDA; 25, 50, and 100 nmol/eye), the number of cells located in the ganglion cell layer (GCL) was reduced, along with thinning of the inner plexiform layer. Intravitreal resveratrol injection (100 nmol/eye) reduced the NMDA (25 and 50 nmol/eye)-induced cell loss in the GCL. The neuroprotective effect of resveratrol was significantly but not completely reversed by compound C (10 nmol/eye). These results suggest that resveratrol dilates retinal arterioles and protects against NMDA-induced retinal neurodegeneration via an AMPK-dependent pathway in rats. Resveratrol may have the potential to slow the onset and progression of diseases associated with retinal ischemia by improving impaired retinal circulation and protecting retinal neuronal cells.


Asunto(s)
N-Metilaspartato , Resveratrol , Células Ganglionares de la Retina , Animales , Masculino , Ratas , Proteínas Quinasas Activadas por AMP/metabolismo , Arteriolas/efectos de los fármacos , N-Metilaspartato/efectos adversos , N-Metilaspartato/farmacología , Ratas Wistar , Resveratrol/farmacología , Retina/metabolismo
6.
Cell Tissue Res ; 390(2): 173-187, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35895162

RESUMEN

Matrix metalloproteinases (MMPs) and tumor necrosis factor (TNF)-α contribute to the pathogenesis of several ocular diseases. Previous studies have shown that MMP-9 activation plays an important role in capillary degeneration in injured retinas. In this study, we aimed to determine the roles of TNF-α in capillary degeneration and MMP-9 activation in the injured retina. In rats, retinal injury was induced by intravitreal injection of N-methyl-D-aspartic acid (NMDA, 200 nmol) at postnatal day 7. We examined (1) the effects of blocking MMP-9 and TNF-α signaling pathway on capillary degeneration, (2) changes in protein levels and distribution of MMP-9 and TNF-α, and (3) the interaction between MMP-9 and TNF-α in regulating the expression level of each protein in retinas of NMDA-injected eyes. Intravitreal injection of GM6001, an MMP inhibitor, or TNF-α neutralizing antibody (anti-TNF-α Ab) attenuated capillary degeneration in retinas of NMDA-injected eyes. Protein levels of TNF-α increased 2 h after NMDA injection, whereas those of MMP-9 increased 4 h after the injection. Anti-TNF-α Ab suppressed activation of MMP-9 in retinas of NMDA-injected eyes, whereas GM6001 diminished the TNF-α protein expression. Incubation of recombinant TNF-α with supernatants of homogenized retina increased protein levels and activity of MMP-9. These results suggest that TNF-α and MMP-9 collaboratively increase their expression levels in the retina following neurodegeneration, thus leading to retinal capillary degeneration. The cooperative interaction between MMP-9 and TNF-α could be involved in the exacerbation of retinal neurovascular degeneration.


Asunto(s)
Metaloproteinasa 9 de la Matriz , Degeneración Retiniana , Ratas , Animales , Metaloproteinasa 9 de la Matriz/metabolismo , N-Metilaspartato/efectos adversos , Factor de Necrosis Tumoral alfa/metabolismo , Animales Recién Nacidos , Inhibidores del Factor de Necrosis Tumoral , Retina/metabolismo , Degeneración Retiniana/patología
7.
Exp Eye Res ; 220: 109107, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35568201

RESUMEN

Na+/K+-ATPase (NKA) plays an important role in ion homeostasis and neurotransmitter uptake. In the retina, multidirectional communications among neurons, glia, and blood vessels (that is, neuro-glio-vascular interaction) are crucial for maintaining tissue homeostasis. We investigated the role of NKA in the elements of neuro-glio-vascular unit in neonatal and adult rat retinas. Male Sprague-Dawley rats (1- and 8-week-old) were injected intravitreally with ouabain (20 nmol/eye), an inhibitor of NKA. Morphological changes in retinal neurons, glia, and blood vessels were examined. The intravitreal injection of ouabain decreased the number of cells in the ganglion cell layer, as well as the thicknesses of the inner plexiform and inner nuclear layers in neonatal and adult rats compared to age-matched controls. The ouabain-induced neuronal cell damage was partially prevented by D-(-)-2-amino-5-phosphonopentanoic acid, an antagonist of N-methyl-D-aspartic acid receptors. In the deep retinal vascular plexus of the ouabain-injected eyes, angiogenesis was delayed in neonatal rats, whereas capillary degeneration occurred in adult rats. The immunoreactivity of glutamine synthetase and vascular endothelial growth factor (VEGF) decreased in the retinas of neonatal and adult rats injected intravitreally with ouabain. The immunoreactivity of glial fibrillary acidic protein was enhanced in the retinas of ouabain-injected adult eyes. After the ouabain injection, CD45-positive leukocytes and Iba1-positive microglia increased in the inner retinal layer of neonatal rats, whereas they increased in the middle retinal layer of adult rats. These results suggest that the inhibition of NKA induces the degeneration of neuronal and vascular cells and alteration of glial cells in both neonatal and adult retinas. In addition to the direct effects of NKA inhibition, the disturbance of retinal glutamate metabolism and decreased VEGF expression may contribute to neurovascular degeneration. The activity of NKA is crucial for maintaining elements of neuro-glio-vascular unit in the retina.


Asunto(s)
Ouabaína , Factor A de Crecimiento Endotelial Vascular , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/farmacología , Animales , Masculino , Neuroglía/metabolismo , Ouabaína/metabolismo , Ouabaína/farmacología , Ratas , Ratas Sprague-Dawley , Retina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
J Pharmacol Sci ; 146(4): 233-243, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34116737

RESUMEN

Retinopathy of prematurity (ROP) is a proliferative retinal vascular disease, initiated by delayed retinal vascular growth after premature birth. In the majority of cases, ROP resolves spontaneously; however, a history of ROP may increase the risk of long-term visual problems. In this study, we evaluated the endothelial function of retinal blood vessels in adult rats with a history of ROP. ROP was induced in rats by subcutaneous injection of a vascular endothelial growth factor receptor tyrosine kinase inhibitor (KRN633) on postnatal day (P) 7 and P8. On P56, vasodilator responses to acetylcholine, GSK1016790A (an activator of transient receptor potential vanilloid 4 channels), NOR3 (a nitric oxide [NO] donor), and salbutamol (a ß2-adrenoceptor agonist) were assessed. Compared to age-matched controls, retinal vasodilator responses to acetylcholine and GSK1016790A were attenuated in P56 rats with a history of ROP. No attenuation of acetylcholine-induced retinal vasodilator response was observed under inhibition of NO synthase. Retinal vasodilator responses to NOR3 and salbutamol were unaffected. These results suggest that the production of and/or release of NO is impaired in retinal blood vessels in adult rats with a history of ROP. A history of ROP might increase the risk of impaired retinal circulation in adulthood.


Asunto(s)
Endotelio Vascular/fisiopatología , Vasos Retinianos/fisiopatología , Retinopatía de la Prematuridad/fisiopatología , Vasodilatación , Acetilcolina/farmacología , Albuterol/farmacología , Animales , Animales Recién Nacidos , Circulación Sanguínea/efectos de los fármacos , Femenino , Leucina/análogos & derivados , Leucina/farmacología , Óxido Nítrico/fisiología , Donantes de Óxido Nítrico/farmacología , Embarazo , Ratas Sprague-Dawley , Sulfonamidas/farmacología , Vasodilatación/efectos de los fármacos
9.
Int J Mol Sci ; 22(9)2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33922757

RESUMEN

Metformin, an anti-hyperglycemic drug of the biguanide class, exerts positive effects in several non-diabetes-related diseases. In this study, we aimed to examine the protective effects of metformin against N-methyl-D-aspartic acid (NMDA)-induced excitotoxic retinal damage in rats and determine the mechanisms of its protective effects. Male Sprague-Dawley rats (7 to 9 weeks old) were used in this study. Following intravitreal injection of NMDA (200 nmol/eye), the number of neuronal cells in the ganglion cell layer and parvalbumin-positive amacrine cells decreased, whereas the number of CD45-positive leukocytes and Iba1-positive microglia increased. Metformin attenuated these NMDA-induced responses. The neuroprotective effect of metformin was abolished by compound C, an inhibitor of AMP-activated protein kinase (AMPK). The AMPK activator, AICAR, exerted a neuroprotective effect in NMDA-induced retinal injury. The MEK1/2 inhibitor, U0126, reduced the neuroprotective effect of metformin. These results suggest that metformin protects against NMDA-induced retinal neurotoxicity through activation of the AMPK and MEK/extracellular signal-regulated kinase (ERK) signaling pathways. This neuroprotective effect could be partially attributable to the inhibitory effects on inflammatory responses.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Metformina/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , N-Metilaspartato/toxicidad , Fármacos Neuroprotectores/farmacología , Enfermedades de la Retina/prevención & control , Animales , Agonistas de Aminoácidos Excitadores/toxicidad , Hipoglucemiantes/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Enfermedades de la Retina/inducido químicamente , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Transducción de Señal
10.
Dev Dyn ; 250(4): 497-512, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33085163

RESUMEN

BACKGROUND: In mice, a tri-layered (superficial, intermediate, and deep) vascular structure is formed in the retina during the third postnatal week. Short-term treatment of newborn mice with vascular endothelial growth factor (VEGF) receptor inhibitors delays the formation of superficial vascular plexus and this allows us to investigate the developmental process of superficial and deep vascular plexuses at the same time. Using this model, we examined the effect of pharmacological depletion of retinal neurons on the formation of superficial and deep vascular plexuses. RESULTS: Neuronal cell loss induced by an intravitreal injection of N-methyl-d-aspartic acid on postnatal day (P) 8 delayed vascular development in the deep layer but not in the superficial layer in mice treated with KRN633, a VEGF receptor inhibitor, on P0 and P1. In KRN633-treated mice, neuronal cell loss decreased the number of vertical sprouts originating from the superficial plexus without affecting the number of angiogenic sprouts growing in front. Neuronal cell loss did not impair networks of fibronectin and astrocytes in the superficial layer. CONCLUSIONS: Our results suggest that inner retinal neurons play a crucial role in forming the deep vascular plexus by directing the sprouts from the superficial blood vessels to the deep layer.


Asunto(s)
Neovascularización Fisiológica , Retina/embriología , Animales , Astrocitos , Femenino , Masculino , Ratones Endogámicos ICR , N-Metilaspartato , Compuestos de Fenilurea , Quinazolinas
11.
Cell Tissue Res ; 382(3): 529-549, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32897421

RESUMEN

Misdirected vascular growth frequently occurs in the neovascular diseases in the retina. However, the mechanisms are still not fully understood. In the present study, we created capillary-free zones in the central and peripheral retinas in neonatal mice by pharmacological blockade of vascular endothelial growth factor (VEGF) signaling. Using this model, we investigated the process and mechanisms of revascularization in the central and peripheral avascular areas. After the completion of a 2-day treatment with the VEGF receptor tyrosine kinase inhibitor KRN633 on postnatal day (P) 4 and P5, revascularization started on P8 in the central avascular area where capillaries had been dropped out. The expression levels of VEGF were higher in the peripheral than in the central avascular area. However, the expansion of the vasculature in the peripheral avascular retina remained suppressed until revascularization had been completed in the central avascular area. Additionally, we found disorganized endothelial cell division, misdirected blood vessels with irregular diameters, and abnormal fibronectin networks at the border of the vascular front and the avascular retina. In the central avascular area, a slight amount of fibronectin as non-vascular component re-formed to provide a scaffold for revascularization. Mechanistic analysis revealed that higher levels of VEGF attenuated the migratory response of endothelial cells without decreasing the proliferative activity. These results suggest that the presence of concentration range of VEGF, which enhances both migration and proliferation of the endothelial cells, and the structurally normal fibronectin network contribute to determine the proper direction of angiogenesis.


Asunto(s)
Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Retina/fisiopatología , Neovascularización Retiniana/fisiopatología , Animales , Animales Recién Nacidos , Ratones
12.
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
13.
Nihon Yakurigaku Zasshi ; 155(2): 81-86, 2020.
Artículo en Japonés | MEDLINE | ID: mdl-32115483

RESUMEN

Because visual information accounts for 80-90% of sensory information that we get from our circumstance, loss of vision seriously diminishes our quality of life. According to a recent epidemiological study, glaucoma is the first, and retinitis pigmentosa (RP) is the second leading causes of acquired blindness in Japan. Degeneration of the retinal ganglion cells (RGC) and photoreceptor cells causes glaucoma and RP, respectively. Intraocular pressure-lowering therapy is an only effective treatment for glaucoma, and the agents that protect RGC directly against glaucomatous injury have not been available yet. In addition, there is no effective treatment for RP at present. microRNAs are a class of small, endogenous, non-coding RNAs comprised of approximately 20 nucleotides. It has been clarified that microRNAs reduces the stability of the target mRNAs and/or repress the translation of the target genes. A single microRNA can affect the transcription of multiple mRNAs, and almost 30% of human genes are thought to be regulated by microRNAs. Therefore, it has been considered that the expression changes of microRNAs are possible to cause various diseases, such as cancer and neurodegenerative diseases. Recently, the expression changes in microRNAs have been reported in the retina of experimental model animals for glaucoma and RP. The expressional changes of microRNAs are suggested to be related with development and progression of glaucoma and RP. Here, we will discuss about the relationship between the expressional changes of microRNAs and neuronal cell death in glaucoma and RP.


Asunto(s)
MicroARNs/genética , Retina/patología , Degeneración Retiniana/genética , Animales , Modelos Animales de Enfermedad , Glaucoma/genética , Glaucoma/patología , Humanos , Degeneración Retiniana/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
14.
Cell Tissue Res ; 379(3): 473-486, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31788758

RESUMEN

An impairment of cellular interactions between the elements of the neurovascular unit contributes to the onset and/or progression of retinal diseases. The present study aims to examine how elements of the neurovascular unit are altered in a rat model of retinopathy of prematurity (ROP). Neonatal rats were treated subcutaneously with the vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor KRN633 (10 mg/kg) on postnatal day (P) 7 and P8 to induce ROP. Morphological assessments were performed of blood vessels, astrocytes and neuronal cells in the retina. Aggressive angiogenesis, tortuous arteries and enlarged veins were observed in the retinal vasculature of KRN633-treated (ROP) rats from P14 to P28, compared to age-matched control (vehicle-treated) animals. Morphological abnormalities in the retinal vasculature showed a tendency toward spontaneous recovery from P28 to P35 in ROP rats. Immunofluorescence staining for glial fibrillary acidic protein and Pax2 (astrocyte markers) revealed that morphological changes to and a reduction in the number of astrocytes occurred in ROP rats. The developmental cell death was slightly accelerated in ROP rats; however, no visible changes in the morphology of retinal layers were observed on P35. The abnormalities in astrocytes might contribute, at least in part, to the formation of abnormal retinal blood vessels and the pathogenesis of ROP.


Asunto(s)
Modelos Animales de Enfermedad , Retina/patología , Neovascularización Retiniana/patología , Retinopatía de la Prematuridad/patología , Animales , Femenino , Compuestos de Fenilurea/farmacología , Embarazo , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Retina/metabolismo , Neovascularización Retiniana/embriología , Neovascularización Retiniana/metabolismo , Retinopatía de la Prematuridad/embriología , Retinopatía de la Prematuridad/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo
15.
Eur J Pharmacol ; 867: 172853, 2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-31836532

RESUMEN

Ca2+-permeable ion channels, such as transient receptor channels, are one of the potential therapeutic targets in cancer. Transient receptor potential vanilloid subtype 4 (TRPV4) is a nonselective cation channel associated with cancer progression. This study investigates the roles of TRPV4 in the pathogenesis of colitis-associated cancer (CAC) in mice. The role of TRPV4 was examined in azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced murine CAC model. The formation of colon tumours induced by AOM/DSS treatment was significantly attenuated in TRPV4-deficient mice (TRPV4KO). TRPV4 was co-localised with markers of angiogenesis and macrophages. AOM/DSS treatment upregulated the expression of CD105, vascular endothelial growth factor receptor 2, and TRPV4 in wildtype, but the upregulation of CD105 was significantly attenuated in TRPV4KO. Bone marrow chimera experiments indicated that TRPV4, expressed in both vascular endothelial cells and bone marrow-derived macrophages, played a significant role in colitis-associated tumorigenesis. There was no significant difference in the population of hematopoietic cells, neutrophils, and monocytes between untreated and AOM/DSS-treated WT and TRPV4KO on flow cytometric analysis. TRPV4 activation by a selective agonist induced TNF-α and CXCL2 release in macrophages. Furthermore, TRPV4 activation enhanced the proliferation of human umbilical vein endothelial cells. These results suggest that TRPV4 expressed in neovascular endothelial cells and bone marrow-derived macrophages contributes to the progression of CAC in mice.


Asunto(s)
Carcinogénesis/patología , Colitis/patología , Neoplasias del Colon/patología , Neoplasias Experimentales/patología , Canales Catiónicos TRPV/metabolismo , Animales , Azoximetano/toxicidad , Carcinogénesis/efectos de los fármacos , Carcinógenos/toxicidad , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL2/metabolismo , Colitis/inducido químicamente , Colon/efectos de los fármacos , Colon/patología , Neoplasias del Colon/inducido químicamente , Sulfato de Dextran/toxicidad , Progresión de la Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Neoplasias Experimentales/inducido químicamente , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/genética
16.
Int J Mol Sci ; 20(19)2019 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-31557901

RESUMEN

Νeuronal and glial cells play an important role in the development of vasculature in the retina. In this study, we investigated whether re-vascularization occurs in retinal neurodegenerative injury models. To induce retinal injury, N-methyl-D-aspartic acid (NMDA, 200 nmol) or kainic acid (KA, 20 nmol) was injected into the vitreous chamber of the eye on postnatal day (P)7. Morphological changes in retinal neurons and vasculature were assessed on P14, P21, and P35. Prevention of vascular growth and regression of some capillaries were observed on P14 in retinas of NMDA- and KA-treated eyes. However, vascular growth and re-vascularization started on P21, and the retinal vascular network was established by P35 in retinas with neurodegenerative injuries. The re-vascularization was suppressed by a two-day treatment with KRN633, an inhibitor of VEGF receptor tyrosine kinase, on P21 and P22. Astrocytes and Müller cells expressed vascular endothelial growth factor (VEGF), and the distribution pattern of VEGF was almost the same between the control and the NMDA-induced retinal neurodegenerative injury model, except for the difference in the thickness of the inner retinal layer. During re-vascularization, angiogenic sprouts from pre-existing blood vessels were present along the network of fibronectins formed by astrocytes. These results suggest that glial cells contribute to angiogenesis in neonatal rat models of retinal neurodegeneration.


Asunto(s)
Degeneración Retiniana/etiología , Degeneración Retiniana/metabolismo , Neovascularización Retiniana/etiología , Neovascularización Retiniana/metabolismo , Animales , Animales Recién Nacidos , Biomarcadores , Modelos Animales de Enfermedad , Técnica del Anticuerpo Fluorescente , Ratas , Degeneración Retiniana/patología , Neovascularización Retiniana/patología , Neuronas Retinianas/metabolismo , Neuronas Retinianas/patología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología
17.
Curr Eye Res ; 44(12): 1360-1368, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31317786

RESUMEN

Purpose: Retinopathy of prematurity (ROP) is characterized by morphological abnormalities in retinal blood vessels, but how an episode of ROP affects vascular function remains to be fully elucidated. The purpose of the present study was to assess the distribution of pericyte/smooth muscle in retinal blood vessels and retinal vasodilator responses in a rat model of ROP.Methods: ROP was induced in rats by the subcutaneous injection of the vascular endothelial growth factor (VEGF) receptor tyrosine kinase inhibitor KRN633 (10 mg/kg) on postnatal day (P) 7 and P8. The distribution of pericyte/smooth muscle in retinal blood vessels was examined on P14 and P35 by immunohistochemistry. Retinal vasodilator responses were assessed on P35 by measuring the diameter of retinal arterioles in fundus images.Results: In retinas of KRN633-treated (ROP) rats, progressive angiogenesis, tortuous arteries, enlarged veins, and enhanced expression of α-smooth muscle actin in pericytes on capillaries and veins were observed on P14. These abnormalities in retinal vasculature showed a tendency to normalize by P35. Vasodilation of retinal arterioles induced by acetylcholine, an endothelium-dependent vasodilator, was smaller in P35 ROP rats than age-matched controls, whereas retinal vasodilator responses to the nitric oxide (NO) donor NOR3 were unaltered.Conclusions: Phenotypic changes in pericytes occur in the ROP model rats and endothelium-dependent vasodilatory mechanisms in retinal blood vessels are impaired. The impaired vasodilator function may contribute to the progression and pathogenesis of ROP.


Asunto(s)
Endotelio Vascular/fisiopatología , Vasos Retinianos/fisiopatología , Retinopatía de la Prematuridad/fisiopatología , Vasodilatación/fisiología , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Inmunohistoquímica , Microscopía Confocal , Ratas , Ratas Sprague-Dawley , Vasos Retinianos/patología , Retinopatía de la Prematuridad/diagnóstico
18.
Int J Mol Sci ; 20(8)2019 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-31010057

RESUMEN

The interactions between neuronal, glial, and vascular cells play a key role in regulating blood flow in the retina. In the present study, we examined the role of the interactions between neuronal and glial cells in regulating the retinal vascular tone in rats upon stimulation of retinal neuronal cells by intravitreal injection of N-methyl-d-aspartic acid (NMDA). The retinal vascular response was assessed by measuring the diameter of the retinal arterioles in the in vivo fundus images. Intravitreal injection of NMDA produced retinal vasodilation that was significantly diminished following the pharmacological inhibition of nitric oxide (NO) synthase (nNOS), loss of inner retinal neurons, or intravitreal injection of glial toxins. Immunohistochemistry revealed the expression of nNOS in ganglion and calretinin-positive amacrine cells. Moreover, glial toxins significantly prevented the retinal vasodilator response induced by intravitreal injection of NOR3, an NO donor. Mechanistic analysis revealed that NO enhanced the production of vasodilatory prostanoids and epoxyeicosatrienoic acids in glial cells in a ryanodine receptor type 1-dependent manner, subsequently inducing the retinal vasodilator response. These results suggest that the NO released from stimulated neuronal cells acts as a key messenger in neuron-glia signaling, thereby causing neuronal activity-dependent and glial cell-mediated vasodilation in the retina.


Asunto(s)
Neuroglía/metabolismo , Neuronas/metabolismo , Vasos Retinianos/metabolismo , Transducción de Señal , Animales , Gangliósidos/metabolismo , Hidroxilaminas , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Masculino , Modelos Biológicos , N-Metilaspartato/metabolismo , N-Metilaspartato/farmacología , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo I/metabolismo , Nitrocompuestos , Prostaglandinas/metabolismo , Ratas Wistar , Vasos Retinianos/patología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/efectos de los fármacos , Vasodilatación/efectos de los fármacos
19.
Exp Eye Res ; 182: 101-108, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30885712

RESUMEN

Interactions between neuronal cells and vascular cells in the retina are critical for maintaining retinal tissue homeostasis. Impairment of cellular interactions contributes to development and progression of retinal diseases. Previous studies demonstrated that neuronal cell damage leads to capillary degeneration in an N-methyl-D-aspartic acid (NMDA)-induced retinal degeneration model. However, the mechanisms underlying this phenomenon are not fully understood. In this study, we examined the possible role of matrix metalloproteinase (MMP)-9 in neuronal cell loss and capillary degeneration in NMDA-treated retinas of neonatal rats. Intravitreal injection of NMDA (50 or 200 nmol) was performed on postnatal day (P) 7 and morphological changes in retinal neurons and vasculature were examined on P14. The MMP inhibitor CP101537 (100 nmol) or vehicle (dimethyl sulfoxide) was intravitreally injected simultaneously with, or 2 days after, NMDA injection. CP101537 protected against neurovascular degeneration in a time-dependent manner as follows: 1) simultaneous injection of CP101537 with NMDA prevented morphological changes in retinal neurons induced by NMDA (50 nmol); and 2) reduction in capillary density and number of vertical sprouts induced by NMDA (200 nmol) was prevented when CP101537 was injected 2 days after NMDA injection. Gelatin zymography and western blot analyses indicated that activity and protein levels of MMP-9 were enhanced from 4 h to 2 days after NMDA injection. Increased activity and protein levels of MMP-9 were suppressed by MMP inhibitors (CP101537 and GM6001). In situ zymography revealed that MMP activity was enhanced throughout the retinal vasculature in NMDA-treated retinas. These results indicate that MMP-9 plays an important role in neurovascular degeneration in the injured retina. Inhibition of MMP-9 may be an effective strategy for preventing and reducing neurovascular degeneration.


Asunto(s)
Capilares/patología , Metaloproteinasa 9 de la Matriz/metabolismo , Degeneración Retiniana/enzimología , Células Ganglionares de la Retina/metabolismo , Vasos Retinianos/patología , Animales , Animales Recién Nacidos , Western Blotting , Capilares/metabolismo , Modelos Animales de Enfermedad , N-Metilaspartato/toxicidad , Ratas Sprague-Dawley , Degeneración Retiniana/inducido químicamente , Degeneración Retiniana/patología , Células Ganglionares de la Retina/patología , Vasos Retinianos/metabolismo
20.
J Pharmacol Sci ; 138(3): 203-208, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30409713

RESUMEN

Pathological retinal angiogenesis contributes to the pathogenesis of several ocular diseases. Valproic acid, a widely used antiepileptic drug, exerts anti-angiogenic effects by inhibiting histone deacetylase (HDAC). Herein, we investigated the effects of valproic acid and vorinostat, a HDAC inhibitor, on pathological retinal angiogenesis in mice with oxygen-induced retinopathy (OIR). OIR was induced in neonatal mice by exposure to 80% oxygen from postnatal day (P) 7 to P10 and to atmospheric oxygen from P10 to P15. Mice were subcutaneously injected with valproic acid, vorinostat, or vehicle once a day from P10 to P14. At P15, retinal neovascular tufts and vascular growth in the central avascular zone were observed in mice with OIR. Additionally, immunoreactivity for phosphorylated ribosomal protein S6 (pS6), an indicator of mammalian target of rapamycin (mTOR) activity, was detected in the neovascular tufts. Both valproic acid and vorinostat reduced the formation of retinal neovascular tuft without affecting vascular growth in the central avascular zone. Valproic acid reduced the pS6 immunoreactivity in neovascular tufts. Given that vascular endothelial growth factor (VEGF) activates mTOR-dependent pathways in proliferating endothelial cells of the neonatal mouse retina, these results suggest that valproic acid suppresses pathological retinal angiogenesis by interrupting VEGF-mTOR pathways.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neovascularización Patológica/prevención & control , Oxígeno/metabolismo , Retina/efectos de los fármacos , Retina/patología , Ácido Valproico/farmacología , Vorinostat/farmacología , Animales , Modelos Animales de Enfermedad , Ratones , Neovascularización Patológica/inducido químicamente , Fosforilación , Retina/metabolismo , Enfermedades de la Retina/sangre , Enfermedades de la Retina/metabolismo , Enfermedades de la Retina/patología , Proteína S6 Ribosómica/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...