Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 43(6): 927-942, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37078291

RESUMEN

BACKGROUND: Endothelial cell activation is tightly controlled by the balance between VEGF (vascular endothelial cell growth factor) and Notch signaling pathway. VEGF destabilizes blood vessels and promotes neovascularization, which are common features of sight-threatening ocular vascular disorders. Here, we show that BCL6B (B-cell CLL/lymphoma 6 member B protein), also known as BAZF, ZBTB28, and ZNF62, plays a pivotal role in the development of retinal edema and neovascularization. METHODS: The pathophysiological physiological role of BCL6B was investigated in cellular and animal models mimicking 2 pathological conditions: retinal vein occlusion and choroidal neovascularization. An in vitro experimental system was used in which human retinal microvascular endothelial cells were supplemented with VEGF. Choroidal neovascularization cynomolgus monkey model was generated to investigate the involvement of BCL6B in the pathogenesis. Mice lacking BCL6B or treated with BCL6B-targeting small-interfering ribose nucleic acid were examined for histological and molecular phenotypes. RESULTS: In retinal endothelial cells, the BCL6B expression level was increased by VEGF. BCL6B-deficient endothelial cells showed Notch signal activation and attenuated cord formation via blockage of the VEGF-VEGFR2 signaling pathway. Optical coherence tomography images showed that choroidal neovascularization lesions were decreased by BCL6B-targeting small-interfering ribose nucleic acid. Although BCL6B mRNA expression was significantly increased in the retina, BCL6B-targeting small-interfering ribose nucleic acid suppressed ocular edema in the neuroretina. The increase in proangiogenic cytokines and breakdown of the inner blood-retinal barrier were abrogated in BCL6B knockout (KO) mice via Notch transcriptional activation by CBF1 (C promotor-binding factor 1) and its activator, the NICD (notch intracellular domain). Immunostaining showed that Müller cell activation, a source of VEGF, was diminished in BCL6B-KO retinas. CONCLUSIONS: These data indicate that BCL6B may be a novel therapeutic target for ocular vascular diseases characterized by ocular neovascularization and edema.


Asunto(s)
Neovascularización Coroidal , Ácidos Nucleicos , Neovascularización Retiniana , Enfermedades Vasculares , Animales , Humanos , Ratones , Neovascularización Coroidal/genética , Neovascularización Coroidal/metabolismo , Células Endoteliales/metabolismo , Macaca fascicularis/metabolismo , Ácidos Nucleicos/metabolismo , Ácidos Nucleicos/uso terapéutico , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Ribosa/metabolismo , Ribosa/uso terapéutico , Enfermedades Vasculares/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Mol Vis ; 29: 188-196, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38222457

RESUMEN

Purpose: To investigate the therapeutic effects of eye drops, namely, timolol maleate, a ß-adrenergic receptor antagonist, and latanoprost, a prostaglandin F2α analog, on retinal edema in a murine retinal vein occlusion (RVO) model. Methods: An RVO model was established using laser-induced RVO in mice, which were administered timolol maleate and latanoprost eye drops several times after venous occlusion. Subsequently, the thickness of the inner nuclear layer (INL) and the expression levels of such genes as Vegf and Atf4, which are stress markers of the endoplasmic reticulum, were examined. Primary human cultured retinal microvascular endothelial cells (HRMECs) were treated with timolol under hypoxic conditions, after which the gene expression pattern was investigated. Importantly, an integrated stress response inhibitor (ISRIB) was used in the RVO model, he known ISRIB, which suppresses the expression of ATF4 in retinal edema. Results: Increased INL thickness was suppressed by timolol eye drops, as were the expressions of Vegf and Atf4, in the RVO model. However, latanoprost eye drops did not induce any change in INL thickness. In HRMECs, hypoxic stress and serum deprivation increased the Vegf and Atf4 expressions; in response, treatment with timolol suppressed the Vegf expression. Furthermore, the ISRIB decreased the Vegf expression pattern and edema formation, which are associated with RVO. Conclusions: These results indicate that timolol eye drops may be a potential option for RVO treatment.


Asunto(s)
Papiledema , Oclusión de la Vena Retiniana , Masculino , Humanos , Ratones , Animales , Timolol/farmacología , Timolol/uso terapéutico , Timolol/metabolismo , Oclusión de la Vena Retiniana/complicaciones , Oclusión de la Vena Retiniana/tratamiento farmacológico , Oclusión de la Vena Retiniana/metabolismo , Soluciones Oftálmicas/uso terapéutico , Latanoprost/farmacología , Latanoprost/metabolismo , Latanoprost/uso terapéutico , Papiledema/tratamiento farmacológico , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Edema/complicaciones
3.
Exp Eye Res ; 228: 109405, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36773739

RESUMEN

This study aimed to determine the role of transient receptor potential vanilloid 4 (TRPV4), a calcium (Ca2+)-permeable cation channel, in the pathophysiology of retinal vascular disease. The retinal vein occlusion (RVO) murine model was created by irradiating retinal veins using lasers. TRPV4 expression and localization were evaluated in RVO mice retinas. In addition, we examined the effects of TRPV4 antagonists (RQ-00317310, HC-067047, GSK2193874, and GSK2798745) on retinal edema, blood flow, and ischemic areas in RVO mice. Furthermore, changes in the retinal expression of tumor necrosis factor (TNF)-α and aquaporin4 (AQP4) by RQ-00317310 were analyzed using Western blot. We also assessed the barrier integrity of epithelial cell monolayers using trans-endothelial electrical resistance (TEER) in Human Retinal Microvascular Endothelial Cells (HRMECs). The expression of TRPV4 was significantly increased and co-localized with glutamine synthetase (GS), a Müller glial marker, in the ganglion cell layer (GCL) of the RVO mice. Moreover, RQ-00317310 administration ameliorated the development of retinal edema and ischemia in RVO mice. In addition, the up regulation of TNF-α and down-regulation of AQP4 were lessened by the treatment with RQ-00317310. Treatment with GSK1016790A, a TRPV4 agonist, increased vascular permeability, while RQ-00317310 treatment decreased vascular endothelial growth factor (VEGF)- or TRPV4-induced retinal vascular hyperpermeability in HRMECs. These findings suggest that TRPV4 plays a role in the development of retinal edema and ischemia. Thus, TRPV4 could be a new therapeutic target against the pathological symptoms of retinal vascular diseases.


Asunto(s)
Papiledema , Enfermedades de la Retina , Canales de Potencial de Receptor Transitorio , Enfermedades Vasculares , Ratones , Humanos , Animales , Permeabilidad Capilar , Canales Catiónicos TRPV/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Canales de Potencial de Receptor Transitorio/farmacología , Células Endoteliales/metabolismo , Papiledema/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Enfermedades de la Retina/metabolismo , Enfermedades Vasculares/metabolismo
4.
FASEB J ; 36(6): e22323, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35485981

RESUMEN

Neovascular glaucoma (NVG) is caused by the formation of new blood vessels in the angle, iris, and cornea in retinal ischemic disease, such as proliferative diabetic retinopathy (PDR) and retinal vein occlusion (RVO), which can reduce the visual acuity. However, the pathophysiological symptoms of NVG are still not well understood because there is no model for the formation of NVG in the angle, iris, and cornea. The aim of this study was to investigate the involvement of NVG during ischemic disease, in a murine model of retinal ischemia. We evaluated the changes of the intraocular pressure (IOP) and pathological symptoms in the anterior eye segment and retina in this model, and the changes in the RNA or protein expression of vascular endothelial growth factor (VEGF) and fibrosis-related factors were analyzed in the retina and cornea by quantitative real-time polymerase chain reaction or western blot, respectively. Furthermore, we examined the changes in IOP after intravitreal injection of an anti-VEGF antibody. First, NVG formed in the retinal ischemic murine model, and the IOP was elevated in mice with NVG formation. Interestingly, VEGF expression was decreased in the retina but increased in the cornea in the murine model of NVG. On the other hand, fibrosis-related factors were increased in the retina and also significantly increased in the cornea in NVG. Moreover, the administration of anti-VEGF antibody immediately after vessel occlusion suppressed the increase in IOP, but administration at 7 days after vessel occlusion accelerated the increase in IOP. These findings suggest that the formation of NVG may be correlated with the pathological symptoms of retinal ischemic disease, via changes in VEGF and fibrosis-related factor expression.


Asunto(s)
Glaucoma Neovascular , Enfermedades de la Retina , Animales , Segmento Anterior del Ojo/irrigación sanguínea , Modelos Animales de Enfermedad , Fibrosis , Glaucoma Neovascular/diagnóstico , Glaucoma Neovascular/etiología , Ratones , Retina , Factor A de Crecimiento Endotelial Vascular/genética
5.
Biol Pharm Bull ; 46(3): 473-481, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36858576

RESUMEN

Macular edema causes vision loss in patients with retinal vein occlusion (RVO) and diabetic macular edema (DME). The intravitreal injection of anti-vascular endothelial growth factor (VEGF) agents is used for treatment; however, this therapy is invasive, and recurrence occurs in some cases. The establishment of a non-invasive treatment would help to solve these problems. Here, we focused on arctigenin, a lignan polyphenol found in burdock sprout, and has effects on inflammatory and microcirculatory when taken orally. We hypothesized that oral intake of arctigenin could be effective against retinal edema in RVO and DME. In this study, the degree of retinal edema by measuring the total retinal thickness using optical coherence tomography (OCT) and the thickness of the inner nuclear layer (INL) by hematoxylin-eosin (H&E) staining were investigated. Oral administration of arctigenin ameliorated retinal edema in an RVO murine model by inhibiting the decrease in occludin and vascular endothelial (VE)-cadherin. Moreover, in retinas with edema, arctigenin suppressed the induction of VEGF, tumor necrosis factor α (TNFα), and matrix metallopeptidase 9 (MMP9). Next, the effects of arctigenin on barrier function were assessed in human retinal microvascular endothelial cells (HRMECs) by measuring the trans-endothelial electrical resistance (TEER) and conducting fluorescein isothiocyanate (FITC)-dextran permeability assays. Arctigenin showed a protective effect against VEGF-induced barrier dysfunction. In addition, arctigenin inhibited the TNFα-mediated activation of the nuclear factor-kappaB (NF-κB)/p38 mitogen-activated protein kinase (MAPK) pathway. These results suggested that oral administration of arctigenin has beneficial effects on retinal edema by inhibiting vascular hyperpermeability in endothelial cells.


Asunto(s)
Retinopatía Diabética , Lignanos , Edema Macular , Papiledema , Oclusión de la Vena Retiniana , Humanos , Animales , Ratones , Células Endoteliales , Microcirculación , Factor de Necrosis Tumoral alfa , Factor A de Crecimiento Endotelial Vascular
6.
FASEB J ; 35(2): e21313, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33484194

RESUMEN

Anti-vascular endothelial growth factor (VEGF) therapies are now the first-line treatment for many ocular diseases, but some patients are non-responders to these therapies. The purpose of this study was to determine whether the level of adiponectin increased the pathogenesis of retinal edema and neovascularization in the retina of progressive ocular vascular diseases. We examined the role played by adiponectin in two types of cells and animal models which are retinal vein occlusion (RVO) and oxygen-induced retinopathy (OIR) mice. Our results showed that an injection of anti-adiponectin antibody ameliorated the retinal edema and ischemia through the depression of the expression level of VEGF-related factors and tight junction-related proteins in the retina of RVO mice. The intravitreal injection of anti-adiponectin antibody also decreased the degree of retinal neovascularization in an OIR mice. In addition, exposure of human retinal microvascular endothelial cells and human brain microvascular pericytes in culture to adiponectin increased both the vascular permeability and neovascularization through the increase of inflammatory factor and the dropout of the pericytes. These findings indicate that adiponectin plays a critical role in retinal edema and neovascularization, and adiponectin is a potential therapeutic target for the treatment of diabetic macular edema, proliferative diabetic retinopathy, and RVO.


Asunto(s)
Adiponectina/metabolismo , Oftalmopatías/metabolismo , Oftalmopatías/patología , Ojo/metabolismo , Ojo/patología , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología , Adiponectina/genética , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Humanos , Masculino , Ratones , Papiledema/metabolismo , Papiledema/patología , Reacción en Cadena en Tiempo Real de la Polimerasa , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/metabolismo , Vasos Retinianos/patología
7.
Exp Eye Res ; 204: 108453, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33503450

RESUMEN

Retinal vein occlusion (RVO) is an intractable eye disease that results in reduced visual acuity, associated with retinal ischemia, hemorrhage, and edema. RVO results in excessive ROS production in the retina, causing inflammation and retinal edema. A free radical scavenger, 4-(4-acetylpiperazin-1-yl)-2-(1H-imidazole-1-yl) aniline (NSP-116), has been reported to demonstrate antioxidative effects and prevent ROS production in the retina. Therefore, NSP-116 may represent a useful drug for treating the pathological symptoms of RVO, such as retinal edema and ischemic symptoms. This study aimed to investigate the effects of NSP-116 in a murine model of RVO. We evaluated the thickness of the retinal layer and the size of the non-perfused area following the oral administration of NSP-116. Moreover, we used western blot analysis to examine the expression levels of vascular endothelial growth factor (VEGF) and tumor necrosis factor (TNF)-α, after NSP-116 administration, and examined the localization of 8-hydroxy-2'-deoxyguanosine (8-OHdG), by immunostaining. The findings indicate that NSP-116 suppressed retinal edema and expansion the non-perfused area by suppressing the increased expression of VEGF, TNF-α, and 8-OHdG in the murine RVO model. In conclusion, the oral administration of NSP-116 may serve as an effective pharmacological treatment for the pathological symptoms of RVO.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Modelos Animales de Enfermedad , Depuradores de Radicales Libres/uso terapéutico , Imidazoles/uso terapéutico , Oclusión de la Vena Retiniana/prevención & control , 8-Hidroxi-2'-Desoxicoguanosina/metabolismo , Administración Oral , Animales , Western Blotting , Angiografía con Fluoresceína , Edema Macular/diagnóstico por imagen , Edema Macular/metabolismo , Edema Macular/fisiopatología , Edema Macular/prevención & control , Ratones , Flujo Sanguíneo Regional/fisiología , Oclusión de la Vena Retiniana/diagnóstico por imagen , Oclusión de la Vena Retiniana/metabolismo , Oclusión de la Vena Retiniana/fisiopatología , Tomografía de Coherencia Óptica , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Exp Eye Res ; 204: 108441, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33453278

RESUMEN

Retinal vein occlusion (RVO) is a vascular disease that represents characteristic retinal hemorrhage and dilated retinal veins. Despite its clinical importance, its pathogenesis remains largely unknown because of limited opportunities to acquire human retinal samples. Therefore, an animal model that reproduces the clinical features of RVO patients is required for further investigation. In this study, we established a pigmented murine RVO model that reproduced characteristic fundus appearances similar to human RVO findings. Retinal edema in this model was observed in both optical coherence tomography and histological analysis, which is a clinically important outcome. With quantitative real-time PCR analysis on retinal samples, we revealed that the mRNA level of vascular endothelial growth factor (VEGF) increased in the retina induced RVO. Moreover, this retinal edema was reduced by intravitreal injection of anti-VEGF antibody. These results were consistent with human clinical knowledge and suggested that this model could be a useful tool for research into new therapeutic approaches.


Asunto(s)
Modelos Animales de Enfermedad , Oclusión de la Vena Retiniana/patología , Animales , Anticuerpos Antiidiotipos/uso terapéutico , Angiografía con Fluoresceína , Humanos , Inmunoglobulina G/inmunología , Edema Macular/diagnóstico , Edema Macular/tratamiento farmacológico , Edema Macular/patología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Oclusión de la Vena Retiniana/diagnóstico por imagen , Oclusión de la Vena Retiniana/tratamiento farmacológico , Tomografía de Coherencia Óptica , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/inmunología
9.
Int J Mol Sci ; 22(16)2021 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-34445595

RESUMEN

Neovascular age-related macular degeneration (nAMD) featuring choroidal neovascularization (CNV) is the principal cause of irreversible blindness in elderly people in the world. Integrated stress response (ISR) is one of the intracellular signals to be adapted to various stress conditions including endoplasmic reticulum (ER) stress. ISR signaling results in the upregulation of activating transcription factor 4 (ATF4), which is a mediator of ISR. Although recent studies have suggested ISR contributes to the progression of some age-related disorders, the effects of ATF4 on the development of CNV remain unclear. Here, we performed a murine model of laser-induced CNV and found that ATF4 was highly expressed in endothelial cells of the blood vessels of the CNV lesion site. Exposure to integrated stress inhibitor (ISRIB) reduced CNV formation, vascular leakage, and the upregulation of vascular endothelial growth factor (VEGF) in retinal pigment epithelium (RPE)-choroid-sclera complex. In human retinal microvascular endothelial cells (HRMECs), ISRIB reduced the level of ATF4 and VEGF induced by an ER stress inducer, thapsigargin, and recombinant human VEGF. Moreover, ISRIB decreased the VEGF-induced cell proliferation and migration of HRMECs. Collectively, our findings showed that pro-angiogenic effects of ATF4 in endothelial cells may be a potentially therapeutic target for patients with nAMD.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Células Endoteliales/patología , Epitelio Pigmentado de la Retina/patología , Factores de Crecimiento Endotelial Vascular/metabolismo , Factor de Transcripción Activador 4/genética , Animales , Neovascularización Coroidal/etiología , Neovascularización Coroidal/metabolismo , Células Endoteliales/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal , Factores de Crecimiento Endotelial Vascular/genética
10.
Mol Vis ; 25: 859-868, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31908404

RESUMEN

Purpose: We investigated the effects of oral and ocular administration of crocetin in a murine retinal vein occlusion (RVO) model. Crocetin is a type of carotenoid contained in the fruit of gardenia (Gardenia jasminoides Ellis) and the stigma of saffron (Crocus staruts L). Methods: This study was performed on a murine RVO model, which was created by laser irradiation of retinal veins. We evaluated the retinal thickness after the oral administration of crocetin (100 mg/kg) 1 and 6 h before laser irradiation, and immediately, 6 h, 12 h, and 18 h after laser irradiation in the murine RVO model. In addition, we measured the retinal layer thickness after administration of crocetin eye drops (0.03% or 0.10%) immediately, 6 h, and 12 h after laser irradiation. Western blotting of retinal tissue was used to determine the expression levels of matrix metalloproteinase (MMP-9), tumor nuclear factor (TNF-α), and occludin after oral administration of crocetin. Results: Oral and ocular administration of crocetin improved retinal edema in the murine RVO model. Crocetin administration statistically significantly suppressed overexpression of MMP-9 and TNF-α, and reversed the reduction of occludin. Conclusions: These findings indicate that crocetin can protect retinal tight junctions by suppressing retinal edema through an anti-inflammatory effect, which suggests that crocetin may be useful for RVO disease.


Asunto(s)
Carotenoides/administración & dosificación , Carotenoides/uso terapéutico , Edema/tratamiento farmacológico , Edema/prevención & control , Retina/patología , Oclusión de la Vena Retiniana/tratamiento farmacológico , Administración Oftálmica , Administración Oral , Animales , Carotenoides/farmacología , Modelos Animales de Enfermedad , Edema/complicaciones , Mediadores de Inflamación/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ocludina/metabolismo , Soluciones Oftálmicas/administración & dosificación , Soluciones Oftálmicas/farmacología , Soluciones Oftálmicas/uso terapéutico , Oclusión de la Vena Retiniana/complicaciones , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Vitamina A/análogos & derivados
11.
Exp Eye Res ; 177: 1-11, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30040948

RESUMEN

To determine the characteristics of the damages of the retinal pigment epithelium (RPE) and photoreceptors of pigmented mice induced by exposure to blue light emitting diode (LED) light, and to determine the mechanisms causing the damages. Exposure to blue LED light for 3 days induced retinal damage, and the characteristics of the damage differed from that induced by white fluorescent light exposure. Ophthalmoscopy showed that blue LED exposure for 3 days induced white spots on the retina, and histological examinations showed materials accumulated at the IS/OS junction of the photoreceptors. The accumulated materials were stained by ionized calcium binding adapter molecule-1 (Iba-1), a marker for macrophages. The debris was also positive for periodic acid-Schiff (PAS). An enlarging the area of RPE was detected just after the blue LED exposure especially around the optic nerve, and this led to a secondary degeneration of the photoreceptors. Exposure of pigmented mice to 3 consecutive days of blue LED light will cause RPE and photoreceptor damage. The damage led to an accumulation of macrophages and drusen-like materials around the outer segments of the photoreceptors. This blue light exposed model may be useful for investigating the pathogenesis of non-exudative age-related macular degeneration.


Asunto(s)
Luz/efectos adversos , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Degeneración Retiniana/patología , Epitelio Pigmentado de la Retina/efectos de la radiación , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Estrés Oxidativo , Retina/fisiopatología , Retina/efectos de la radiación , Drusas Retinianas/patología
12.
J Pharmacol Sci ; 137(2): 129-136, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29983234

RESUMEN

Rho-associated coiled-coil containing protein kinase (ROCK) inhibitors are used to treat glaucoma patients and have protective effects on ischemic states. However, it is poorly understood how the ROCK pathway affects the pathological signs of retinal vein occlusion (RVO). In this study, we evaluated the effects of ripasudil, a ROCK inhibitor, on a murine RVO model. In vivo, RVO was induced by retinal vein laser irradiation in mice, and evaluated with ripasudil. In vitro, the effects of ripasudil were examined on tight junction protein integrity in human retinal microvascular endothelial cells (HRMECs). Moreover, we investigated the expression level of the phosphorylated myosin phosphatase target protein (MYPT)-1 after administration of ripasudil. Ripasudil significantly prevented deterioration, such as retinal edema, reduced the size of the nonperfusion area, and improved retinal blood flow. Ripasudil treatment inhibited disintegration of ZO-1 in HRMECs. Administration of ripasudil suppressed retinal phosphorylation of MYPT-1 in a murine RVO model. These findings indicate that ripasudil might be as a possible therapeutic agent for RVO.


Asunto(s)
Isquemia/tratamiento farmacológico , Isoquinolinas/administración & dosificación , Isoquinolinas/farmacología , Papiledema/tratamiento farmacológico , Oclusión de la Vena Retiniana/tratamiento farmacológico , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Inyecciones Intravítreas , Masculino , Ratones Endogámicos , Fosfatasa de Miosina de Cadena Ligera/genética , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación/efectos de los fármacos , Retina/citología , Oclusión de la Vena Retiniana/genética , Oclusión de la Vena Retiniana/metabolismo , Vasos Retinianos , Proteínas de Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
13.
J Pharmacol Sci ; 134(2): 86-92, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28619445

RESUMEN

Kallidinogenase has been used to treat retinal vein occlusion (RVO) in patients, although there are no evidences on the effects of kallidinogenase on the retinal edema and the non-perfused areas in eyes with a RVO. We have established a murine RVO model with retinal edema and non-perfused areas. The purpose of this study was to evaluate the effects of kallidinogenase on the retinal edema and size of the non-perfused areas in the mouse RVO model. We evaluated the thickness of the retinal layers and size of the non-perfused areas, and the blood flow by laser speckle flowgraphy in RVO model. The effects of an intravenous injection of kallidinogenase on the retinal edema and size of the non-perfused areas were determined. In addition, the expressions of phosphorylated protein kinase B (Akt) and endothelial nitric oxide synthase (eNOS) were measured by Western blotting. Our results showed that kallidinogenase reduced the degree of retinal edema and size of the non-perfused areas by an increase in the blood flow in RVO model. Kallidinogenase also increased the levels of phosphorylated Akt and eNOS. These findings indicate that kallidinogenase acted through Akt/eNOS-dependent phosphorylation. Thus, kallidinogenase should be considered as a possible therapeutic agent for RVO patients.


Asunto(s)
Calicreínas/uso terapéutico , Papiledema/tratamiento farmacológico , Oclusión de la Vena Retiniana/tratamiento farmacológico , Animales , Humanos , Masculino , Ratones , Óxido Nítrico Sintasa de Tipo III/metabolismo , Papiledema/metabolismo , Papiledema/patología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Flujo Sanguíneo Regional , Oclusión de la Vena Retiniana/metabolismo , Oclusión de la Vena Retiniana/patología
14.
Sci Rep ; 14(1): 9700, 2024 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-38678148

RESUMEN

Ocular abnormal angiogenesis and edema are featured in several ocular diseases. S1P signaling via S1P1 likely is part of the negative feedback mechanism necessary to maintain vascular health. In this study, we conducted pharmacological experiments to determine whether ASP4058, a sphingosine 1-phosphate receptor 1/5 (S1P1/5) agonist, is useful in abnormal vascular pathology in the eye. First, human retinal microvascular endothelial cells (HRMECs) were examined using vascular endothelial growth factor (VEGF)-induced cell proliferation and hyperpermeability. ASP4058 showed high affinity and inhibited VEGF-induced proliferation and hyperpermeability of HRMECs. Furthermore, S1P1 expression and localization changes were examined in the murine laser-induced choroidal neovascularization (CNV) model, a mouse model of exudative age-related macular degeneration, and the efficacy of ASP4058 was verified. In the CNV model mice, S1P1 tended to decrease in expression immediately after laser irradiation and colocalized with endothelial cells and Müller glial cells. Oral administration of ASP4058 also suppressed vascular hyperpermeability and CNV, and the effect was comparable to that of the intravitreal administration of aflibercept, an anti-VEGF drug. Next, efficacy was also examined in a retinal vein occlusion (RVO) model in which retinal vascular permeability was increased. ASP4058 dose-dependently suppressed the intraretinal edema. In addition, it suppressed the expansion of the perfusion area observed in the RVO model. ASP4058 also suppressed the production of VEGF in the eye. Collectively, ASP4058 can be a potential therapeutic agent that normalizes abnormal vascular pathology, such as age-related macular degeneration and RVO, through its direct action on endothelial cells.


Asunto(s)
Neovascularización Coroidal , Modelos Animales de Enfermedad , Animales , Humanos , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Ratones , Receptores de Esfingosina-1-Fosfato/metabolismo , Receptores de Esfingosina-1-Fosfato/agonistas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proliferación Celular/efectos de los fármacos , Ratones Endogámicos C57BL , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/metabolismo , Masculino
15.
Front Immunol ; 14: 1116238, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36891311

RESUMEN

Background: Adjuvants are chemical or biological materials that enhance the efficacy of vaccines. A-910823 is a squalene-based emulsion adjuvant used for S-268019-b, a novel vaccine against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that is currently in clinical development. Published evidence has demonstrated that A-910823 can enhance the induction of neutralizing antibodies against SARS-CoV-2 in humans and animal models. However, the characteristics and mechanisms of the immune responses induced by A-910823 are not yet known. Methods and Results: To characterize A-910823, we compared the adaptive immune response profile enhanced by A-910823 with that of other adjuvants (AddaVax, QS21, aluminum salt-based adjuvants, and empty lipid nanoparticle [eLNP]) in a murine model. Compared with other adjuvants, A-910823 enhanced humoral immune responses to an equal or greater extent following potent T follicular helper (Tfh) and germinal center B (GCB) cell induction, without inducing a strong systemic inflammatory cytokine response. Furthermore, S-268019-b containing A-910823 adjuvant produced similar results even when given as a booster dose following primary administration of a lipid nanoparticle-encapsulated messenger RNA (mRNA-LNP) vaccine. Preparation of modified A-910823 adjuvants to identify which components of A-910823 play a role in driving the adjuvant effect and detailed evaluation of the immunological characteristics induced by each adjuvant showed that the induction of humoral immunity and Tfh and GCB cell induction in A-910823 were dependent on α-tocopherol. Finally, we revealed that the recruitment of inflammatory cells to the draining lymph nodes and induction of serum cytokines and chemokines by A-910823 were also dependent on the α-tocopherol component. Conclusions: This study demonstrates that the novel adjuvant A-910823 is capable of robust Tfh cell induction and humoral immune responses, even when given as a booster dose. The findings also emphasize that α-tocopherol drives the potent Tfh-inducing adjuvant function of A-910823. Overall, our data provide key information that may inform the future production of improved adjuvants.


Asunto(s)
COVID-19 , Inmunidad Humoral , Humanos , Animales , Ratones , Células T Auxiliares Foliculares , alfa-Tocoferol/farmacología , Escualeno/farmacología , Emulsiones , SARS-CoV-2 , Adyuvantes Inmunológicos/farmacología , Adyuvantes Farmacéuticos
16.
Invest Ophthalmol Vis Sci ; 63(8): 14, 2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35822950

RESUMEN

Purpose: Semaphorin 3A (Sema3A) is a promising therapeutic target for macular edema in age-related macular degeneration, diabetic retinopathy, and retinal vein occlusion (RVO). Anti-vascular endothelial growth factors (anti-VEGFs) are the current standard of care for many retinal diseases. This study investigated the Sema3A neutralizing antibody BI-X and/or anti-VEGF therapy (aflibercept) in an RVO mouse model. Treatment efficacy was examined and grouped by timing subsequent to the RVO mouse model induction: efficacy against the onset of intraretinal edema 1 day postinduction and protective effects at 7 days postinduction. Methods: We examined the changes in expression of Sema3A in the retina of an RVO mouse model. In addition, changes in expression of tumor necrosis factor (TNF)-α and semaphorin-related proteins (neuropilin-1 and plexin A1) in the retina upon treatment were analyzed by Western blotting. The effects of BI-X and/or aflibercept were evaluated using measures of retinal edema, blood flow, and thinning of the inner nuclear layer. Results: Induction of vein occlusion in the RVO mouse model significantly increased Sema3A expression in the retina, particularly in the inner nuclear layer. BI-X was effective as a monotherapy and in combination with anti-VEGF therapy, demonstrating a beneficial effect on intraretinal edema and retinal blood flow. Moreover, in the RVO mouse model, BI-X monotherapy normalized the changes in expression of TNF-α and semaphorin-related proteins. Conclusions: These findings support targeting Sema3A to treat intraretinal edema and retinal ischemia.


Asunto(s)
Edema Macular , Enfermedades de la Retina , Oclusión de la Vena Retiniana , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Modelos Animales de Enfermedad , Inyecciones Intravítreas , Edema Macular/tratamiento farmacológico , Masculino , Ratones , Retina/patología , Enfermedades de la Retina/patología , Oclusión de la Vena Retiniana/metabolismo , Semaforina-3A/metabolismo
17.
Artículo en Inglés | MEDLINE | ID: mdl-32328137

RESUMEN

Acupuncture is used to treat a wide variety of eye diseases, although there is little evidence about the effects of acupuncture treatment and the mechanisms responsible for them. Foot acupuncture treatment has effects in both mice and humans. The purpose of this study was to investigate the effects of acupuncture treatment on ocular blood flow in mice. We evaluated ocular blood flow in C57BL/6J mice after foot acupuncture treatment using laser speckle flowgraphy. The mean blur rate, which is an index of blood flow velocity, was increased in the foot acupuncture group. Our results showed that, after 3 minutes' foot acupuncture, ocular blood flow was significantly increased in both the blood vessels and tissue of the eye in C57BL/6J mice. Thus, performing acupuncture in mice might help to determine its effects. Furthermore, acupuncture is considered to be a possible treatment for ocular disease.

18.
Sci Rep ; 10(1): 11842, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32678228

RESUMEN

Progranulin is a secreted growth factor associated with multiple physiological functions in ischemic pathophysiology. However, it is still not fully understood how progranulin is involved in ischemic lesion and cardiac remodeling after myocardial infarction (MI). In this study, we investigated the effects of progranulin on myocardial ischemia and reperfusion injury. We investigated progranulin expression using Western blotting and immunostaining after permanent left coronary artery (LCA) occlusion in mice. Infarct size and the number of infiltrating neutrophils were measured 24 h after permanent LCA occlusion. Recombinant mouse progranulin was administered before LCA occlusion. In addition, we evaluated cardiac function using cardiac catheterization and echocardiography, and fibrosis size by Masson's trichrome staining after myocardial ischemia/reperfusion in rabbits. Recombinant human progranulin was administered immediately after induction of reperfusion. Progranulin expression increased in the myocardial ischemic area 1, 3, and 5 days after permanent LCA occlusion in mice. The administration of recombinant mouse progranulin significantly attenuated infarct size and infiltrating neutrophils 24 h after permanent LCA occlusion in mice. We also found that administration of recombinant human progranulin ameliorated the deterioration of cardiac dysfunction and fibrosis after myocardial ischemia/reperfusion in rabbits. These findings suggest that progranulin may protect myocardial ischemia/reperfusion injury.


Asunto(s)
Cardiotónicos/farmacología , Fibrosis Endomiocárdica/tratamiento farmacológico , Infarto del Miocardio/tratamiento farmacológico , Isquemia Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Progranulinas/farmacología , Animales , Trastornos Cerebrovasculares/cirugía , Vasos Coronarios/cirugía , Modelos Animales de Enfermedad , Ecocardiografía , Fibrosis Endomiocárdica/diagnóstico por imagen , Fibrosis Endomiocárdica/patología , Humanos , Masculino , Ratones , Ratones Endogámicos ICR , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Isquemia Miocárdica/diagnóstico por imagen , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/diagnóstico por imagen , Daño por Reperfusión Miocárdica/patología , Infiltración Neutrófila , Neutrófilos , Conejos , Proteínas Recombinantes/farmacología , Resultado del Tratamiento
19.
Invest Ophthalmol Vis Sci ; 60(6): 1943-1952, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31050722

RESUMEN

Purpose: Ocular angiogenesis, including retinopathy of prematurity, diabetic retinopathy, and exudative age-related macular degeneration, are closely related to oxidative stress. Many reports have shown that the cellular protective mechanism against oxidative stress and inflammatory response has nuclear factor-erythroid 2-related factor-2 (Nrf2) activity. The aim of this study was to investigate the effectiveness and mechanism of Nrf2 activation in treating the ocular diseases with abnormal vessels. Methods: The effects of Nrf2 activators, bardoxolone methyl (BARD) and RS9, were evaluated against vascular endothelial growth factor (VEGF)-induced cell migration in human retinal microvascular endothelial cells (HRMECs). We measured the expression of the Nrf2 target genes, Ho-1 and Nqo-1 mRNA, in mouse retinas after a single injection of BARD and RS9. The effects and mechanisms of RS9 against retinal angiogenesis were evaluated using an oxygen-induced retinopathy (OIR) model in mice. Moreover, the effect of RS9 against choroidal neovascularization (CNV) was evaluated in a laser-induced CNV monkey model. Results: Both BARD and RS9 decreased VEGF-induced cell migration, and significantly increased Ho-1 mRNA expression; however, only RS9 significantly increased Nqo-1 mRNA. RS9 decreased retinal neovascularization through suppressing VEGF expression and increasing Nrf2, HO-1, platelet-derived growth factor receptor (PDGFR)-ß, and tight junction proteins in OIR murine retinas. Furthermore, RS9 showed a tendency toward decreasing CNV lesions, and improved vascular leakage in a CNV monkey model. Conclusions: These data indicate that a Nrf2 activator might be a candidate for treatment of ocular diseases characterized by pathophysiological angiogenesis and hyperpermeability.


Asunto(s)
Neovascularización Coroidal/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/metabolismo , Ácido Oleanólico/análogos & derivados , Neovascularización Retiniana/tratamiento farmacológico , Vasos Retinianos/metabolismo , Triterpenos/farmacología , Animales , Permeabilidad de la Membrana Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Neovascularización Coroidal/metabolismo , Neovascularización Coroidal/patología , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Angiografía con Fluoresceína , Fondo de Ojo , Humanos , Immunoblotting , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Ácido Oleanólico/farmacología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Vasos Retinianos/patología , Tomografía de Coherencia Óptica
20.
Invest Ophthalmol Vis Sci ; 59(11): 4701-4713, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30267092

RESUMEN

Purpose: To determine the relationship between retinal morphologic changes and molecules involved in the changes after anti-VEGF treatment in the retina of retinal vein occlusion (RVO) murine model. Methods: The studies were performed on murine RVO model created by laser irradiation of retinal veins. The site of VEGF expression was determined by immunostaining, and the retinal thickness was measured in the images obtained by optical coherence tomography. The levels of VEGF-related and inflammatory factors after an intravitreal injection of anti-VEGF antibody immediately or 7 days after laser irradiation were determined by Western blotting. Results: The level of VEGF increased in all retinal layers 1 day after laser irradiation, and expression was higher in the partially perfused areas than in the completely nonperfused areas. In eyes with high expression level of VEGF, early administration of anti-VEGF antibody reduced the retinal thickness, and expressions of VEGF and inflammatory factors returned to normal levels. However, the levels of phosphorylated protein kinase B (AKT), extracellular signal-regulated kinase 1 and 2 (ERK1/2), and endothelial nitric oxide synthase (eNOS) were increased by early administration of anti-VEGF antibody. On the other hand, in eyes with low concentration of VEGF, late injection of anti-VEGF antibody induced retinal thinning and the concentrations of phosphorylated AKT, ERK1/2, and eNOS were lower than that in normal group. Furthermore, anti-VEGF antibody lessened the reduction of aquaporin-4. Conclusions: These findings indicate that the effect of anti-VEGF antibody is most likely dependent on its effect on the intraocular VEGF levels.


Asunto(s)
Papiledema/fisiopatología , Retina/fisiopatología , Oclusión de la Vena Retiniana/fisiopatología , Factor A de Crecimiento Endotelial Vascular/fisiología , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Western Blotting , Modelos Animales de Enfermedad , Electroforesis en Gel de Poliacrilamida , Angiografía con Fluoresceína , Inyecciones Intravítreas , Flujometría por Láser-Doppler , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Papiledema/diagnóstico , Papiledema/tratamiento farmacológico , Papiledema/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Retina/metabolismo , Oclusión de la Vena Retiniana/diagnóstico , Oclusión de la Vena Retiniana/tratamiento farmacológico , Oclusión de la Vena Retiniana/metabolismo , Tomografía de Coherencia Óptica , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA