Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 785
Filtrar
1.
Theranostics ; 14(13): 4983-5000, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39267779

RESUMEN

Rationale: Neovascular ocular diseases (NODs) represent the leading cause of visual impairment globally. Despite significant advances in anti-angiogenic therapies targeting vascular endothelial growth factor (VEGF), persistent challenges remain prevalent. As a proof-of-concept study, we herein demonstrate the effectiveness of targeted degradation of VEGF with bispecific aptamer-based lysosome-targeting chimeras (referred to as VED-LYTACs). Methods: VED-LYTACs were constructed with three distinct modules: a mannose-6-phosphate receptor (M6PR)-binding motif containing an M6PR aptamer, a VEGF-binding module with an aptamer targeting VEGF, and a linker essential for bridging and stabilizing the two-aptamer structure. The degradation efficiency of VED-LYTACs via the autophagy-lysosome system was examined using an enzyme-linked immunosorbent assay (ELISA) and immunofluorescence staining. Subsequently, the anti-angiogenic effects of VED-LYTACs were evaluated using in vitro wound healing assay, tube formation assay, three-dimensional sprouting assay, and ex vivo aortic ring sprouting assay. Finally, the potential therapeutic effects of VED-LYTACs on pathological retinal neovascularization and vascular leakage were tested by employing mouse models of NODs. Results: The engineered VED-LYTACs promote the interaction between M6PR and VEGF, consequently facilitating the translocation and degradation of VEGF through the lysosome. Our data show that treatment with VED-LYTACs significantly suppresses VEGF-induced angiogenic activities both in vitro and ex vivo. In addition, intravitreal injection of VED-LYTACs remarkably ameliorates abnormal vascular proliferation and leakage in mouse models of NODs. Conclusion: Our findings present a novel strategy for targeting VEGF degradation with an aptamer-based LYTAC system, effectively ameliorating pathological retinal angiogenesis. These results suggest that VED-LYTACs have potential as therapeutic agents for managing NODs.


Asunto(s)
Aptámeros de Nucleótidos , Lisosomas , Neovascularización Retiniana , Factor A de Crecimiento Endotelial Vascular , Animales , Aptámeros de Nucleótidos/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ratones , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/metabolismo , Humanos , Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Inhibidores de la Angiogénesis/farmacología , Angiogénesis
2.
BMC Ophthalmol ; 24(1): 385, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39218852

RESUMEN

BACKGROUND: Rickettsial disease has been commonly associated with retinitis, retinal vasculitis, and optic nerve involvement, but the development of retinal neovascularization has been very rarely reported. We herein describe a case of rickettsial retinitis complicated with the development of sea-fan retinal neovascularization documented with multimodal imaging, including fundus photography, SS-OCT, fluorescein angiography, and SS-OCT angiography. CASE PRESENTATION: A 26-year-old female with a history of fever one week earlier presented with sudden decreased vision in the left eye. Best-corrected visual acuity (BCVA) was 20/2000 and the patient was diagnosed with rickettsial retinitis along the superotemporal retinal vascular arcade associated with serous retinal detachment and retinal hard exudates. The indirect immunofluorescence test was positive for Rickettsia conorii, and the patient was treated with oral doxycycline (200 mg/day) and oral prednisone (0.75 mg/kg/day, with gradual tapering). Four weeks after presentation, the retinal infiltrate and associated serous retinal detachment had resolved, but retinal hard exudates had increased. A large sea-fan preretinal fibrovascular neovascularization became apparent along the superotemporal retinal vascular arcade, but there was no associated retinal ischemia on fluorescein angiography. The patient received an adjunctive single intravitreal injection of 1.25 bevacizumab. Sequential follow-up examinations showed shrinking of sea-fan retinal neovascularization, a complete resolution of retinal hard exudates, and the development of a self-limited vitreous hemorrhage. On last follow-up, 30 months after intravitreal bevacizumab injection, BCVA was 20/25. CONCLUSION: Patients with rickettsial retinitis may develop a sea-fan retinal neovascularization, with subsequent vitreous hemorrhage, putatively through inflammatory mechanisms. Multimodal imaging including OCT, fluorescein angiography, and OCT-angiography, is highly useful for accurate diagnosis and reliable monitoring of the evolution of retinitis, retinal neovascularization, and other retinal changes. The use of a combination therapy with oral doxycycline and corticosteroids and intravitreal anti-VEGF can improve outcomes.


Asunto(s)
Angiografía con Fluoresceína , Neovascularización Retiniana , Retinitis , Humanos , Femenino , Adulto , Neovascularización Retiniana/diagnóstico , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/etiología , Retinitis/diagnóstico , Retinitis/microbiología , Retinitis/tratamiento farmacológico , Angiografía con Fluoresceína/métodos , Tomografía de Coherencia Óptica/métodos , Infecciones Bacterianas del Ojo/diagnóstico , Infecciones Bacterianas del Ojo/microbiología , Infecciones Bacterianas del Ojo/tratamiento farmacológico , Infecciones por Rickettsia/diagnóstico , Infecciones por Rickettsia/complicaciones , Infecciones por Rickettsia/tratamiento farmacológico , Infecciones por Rickettsia/microbiología , Rickettsia conorii , Fondo de Ojo , Agudeza Visual , Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/administración & dosificación , Inyecciones Intravítreas , Doxiciclina/uso terapéutico , Bevacizumab/uso terapéutico
3.
Invest Ophthalmol Vis Sci ; 65(10): 5, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39093298

RESUMEN

Purpose: Retinal neovascularization is a significant feature of advanced age-related macular degeneration (AMD) and a major cause of blindness in patients with AMD. However, the underlying mechanism of this pathological neovascularization remains unknown. Iron metabolism has been implicated in various biological processes. This study was conducted to investigate the effects of iron metabolism on retinal neovascularization in neovascular AMD (nAMD). Methods: C57BL/6J and very low-density lipoprotein receptor (VLDLR) knockout (Vldlr-/-) mice, a murine model of nAMD, were used in this study. Bulk-RNA sequencing was used to identify differentially expressed genes. Western blot analysis was performed to test the expression of proteins. Iron chelator deferiprone (DFP) was administrated to the mice by oral gavage. Fundus fluorescein angiography was used to evaluate retinal vascular leakage. Immunofluorescence staining was used to detect macrophages and iron-related proteins. Results: RNA sequencing (RNA-seq) results showed altered transferrin expression in the retina and RPE of Vldlr-/- mice. Disrupted iron homeostasis was observed in the retina and RPE of Vldlr-/- mice. DFP mitigated iron overload and significantly reduced retinal neovascularization and vascular leakage. In addition, DFP suppressed the inflammation in Vldlr-/- retinas. The reduced signals of macrophages were observed at sites of neovascularization in the retina and RPE of Vldlr-/- mice after DFP treatment. Further, the IL-6/JAK2/STAT3 signaling pathway was activated in the retina and RPE of Vldlr-/- mice and reversed by DFP treatment. Conclusions: Disrupted iron metabolism may contribute to retinal neovascularization in nAMD. Restoring iron homeostasis by DFP could be a potential therapeutic approach for nAMD.


Asunto(s)
Deferiprona , Modelos Animales de Enfermedad , Homeostasis , Quelantes del Hierro , Hierro , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Retiniana , Animales , Deferiprona/farmacología , Deferiprona/uso terapéutico , Quelantes del Hierro/farmacología , Quelantes del Hierro/uso terapéutico , Ratones , Hierro/metabolismo , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/etiología , Neovascularización Retiniana/patología , Angiografía con Fluoresceína , Receptores de LDL/genética , Receptores de LDL/metabolismo , Western Blotting , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/patología , Degeneración Macular Húmeda/tratamiento farmacológico , Degeneración Macular Húmeda/metabolismo , Factor de Transcripción STAT3/metabolismo , Masculino
4.
Cells ; 13(16)2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39195259

RESUMEN

Retinopathy of prematurity (ROP) has a dual-phase disease pathology; in phase 1, hyperoxia-induced vaso-obliteration occurs in the retinal vasculature due to increased oxidative stress (OS) and inflammation, followed by phase 2, where hypoxia increases the overproduction of growth factors, inducing retinal neovascularization. Toll-like receptor 2 and -4 (TLR2 and TLR4) overactivation, hyper-inflammation, macrophages, and neutrophil infiltration contribute to the developing ROP. AVR-121 and AVR-123 are novel classes of small-molecule dual inhibitors of TLR2/4 tested in a human leukemia monocytic cell line (THP-1) and cord-blood-derived mononuclear cells (CBMCs). Both compounds inhibited TLR2/4 signaling-related inflammatory cytokines in THP-1 cells and inhibited VEGF-induced neovascularization in human retinal endothelial cells (HRECs), which are hallmarks of ROP. In an oxygen-induced retinopathy (OIR) murine model, the intraperitoneal injection of AVR-123 in the hyperoxia phase (P7-P12) or a nanosuspension eyedrop of AVR-123 in the hypoxic phase (P12-P17) significantly reduced vaso-obliteration, angiogenesis, and inflammatory cytokine profiles while not inhibiting the necessary growth factor VEGF in the juvenile mouse eyes. The results are consistent with our hypothesis that targeting the dual TLR2/4 pathway will reduce inflammation, angiogenesis, and vaso-obliteration in vitro and in vivo and reduce cytotoxic immune cells. AVR-123 has the potential to be developed as a therapy for ROP.


Asunto(s)
Inhibidores de la Angiogénesis , Antiinflamatorios , Modelos Animales de Enfermedad , Oxígeno , Retinopatía de la Prematuridad , Animales , Ratones , Humanos , Oxígeno/metabolismo , Retinopatía de la Prematuridad/patología , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/metabolismo , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/uso terapéutico , Antiinflamatorios/farmacología , Ratones Endogámicos C57BL , Neovascularización Retiniana/patología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/tratamiento farmacológico , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/antagonistas & inhibidores , Citocinas/metabolismo , Hiperoxia/complicaciones , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 2/antagonistas & inhibidores , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Bibliotecas de Moléculas Pequeñas/farmacología
5.
J Control Release ; 373: 224-239, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39002796

RESUMEN

Intravitreal injection of biodegradable implant drug carriers shows promise in reducing the injection frequency for neovascular retinal diseases. However, current intravitreal ocular devices have limitations in adjusting drug release rates for individual patients, thereby affecting treatment effectiveness. Accordingly, we developed mesoporous silica nanoparticles (MSNs) featuring a surface that reverse its charge in response to reactive oxygen species (ROS) for efficient delivery of humanin peptide (HN) to retinal epithelial cells (ARPE-19). The MSN core, designed with a pore size of 2.8 nm, ensures a high HN loading capacity 64.4% (w/w). We fine-tuned the external surface of the MSNs by incorporating 20% Acetyl-L-arginine (Ar) to create a partial positive charge, while 80% conjugated thioketal (TK) methoxy polyethylene glycol (mPEG) act as ROS gatekeeper. Ex vivo experiments using bovine eyes revealed the immobilization of Ar-MSNs-TK-PEG (mean zeta potential: 2 mV) in the negatively charged vitreous. However, oxidative stress reversed the surface charge to -25 mV by mPEG loss, facilitating the diffusion of the nanoparticles impeded with HN. In vitro studies showed that ARPE-19 cells effectively internalize HN-loaded Ar-MSNs-TK, subsequently releasing the peptide, which offered protection against oxidative stress-induced apoptosis, as evidenced by reduced TUNEL and caspase3 activation. The inhibition of retinal neovascularization was further validated in an in vivo oxygen-induced retinopathy (OIR) mouse model.


Asunto(s)
Nanopartículas , Especies Reactivas de Oxígeno , Neovascularización Retiniana , Dióxido de Silicio , Animales , Dióxido de Silicio/química , Especies Reactivas de Oxígeno/metabolismo , Humanos , Nanopartículas/química , Bovinos , Neovascularización Retiniana/tratamiento farmacológico , Línea Celular , Porosidad , Sistemas de Liberación de Medicamentos , Portadores de Fármacos/química , Ratones , Polietilenglicoles/química , Ratones Endogámicos C57BL , Apoptosis/efectos de los fármacos , Enfermedades de la Retina/tratamiento farmacológico
6.
Sci Rep ; 14(1): 15647, 2024 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-38977744

RESUMEN

This study aimed to determine whether the state of retinal vascularization after anti-vascular endothelial growth factor (anti-VEGF) injection can help predict the risk of reactivated retinopathy of prematurity (ROP) requiring treatment and whether repeated ranibizumab injection will be effective in such cases. We retrospectively reviewed 24 infants (43 eyes) who received ranibizumab monotherapy between January 2021 and December 2022. All eyes were classified as having non-retreated ROP or retreated ROP. The state of ROP at the time of treatment, the time required for resolution of plus disease, and the extent of vascularization at 4 and 8 weeks after treatment were analyzed. Extent of temporal retinal vascularization was measured with serial fundus images using disc-fovea distance (DF) unit and disc diameter (DD). Reactivated ROP requiring treatment occurred in six infants (25.0%) and ten eyes (23.3%) after ranibizumab treatment. The mean retreatment interval was 9.0 ± 3.3 weeks (range 4-16). In the retreated ROP group, the time required for the resolution of plus disease after primary injection was longer compared to the control group (13.3 days vs 5.2 days), with a mean ROP regression time of 3.4 weeks. All eyes in the retreated ROP showed retinal vascularization < 0.5 DF from the original site at 4 weeks after injection. In 90% of cases with retreated ROP, the extent of vascularization at 8 weeks after injection was within 1 DF from the original ROP site, and all cases showed reactivation in the posterior Zone II area. The extent of retinal neovascularization in the retreated group was an average of 0.7 DD (vs 1.7 DD) and 1.3 DD (vs 3.3 DD) at 4 and 8 weeks after injection, respectively. After ranibizumab retreatment, only one reactivated case with vitreous traction progressed to focal retinal detachment, while all other cases regressed with peripheral vascular development. The continuation of delayed retinal blood vessel development after ≥ 8 weeks may indicate a high likelihood of reactivated ROP requiring treatment. In the absence of vitreous traction, ranibizumab reinjection is likely to be effective in treating reactivated ROP requiring treatment.


Asunto(s)
Inhibidores de la Angiogénesis , Ranibizumab , Retinopatía de la Prematuridad , Humanos , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/patología , Ranibizumab/administración & dosificación , Ranibizumab/uso terapéutico , Masculino , Femenino , Recién Nacido , Estudios Retrospectivos , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/uso terapéutico , Resultado del Tratamiento , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/patología , Inyecciones Intravítreas , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Lactante , Recien Nacido Prematuro
7.
J Nanobiotechnology ; 22(1): 354, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38902775

RESUMEN

Fundus neovascularization diseases are a series of blinding eye diseases that seriously impair vision worldwide. Currently, the means of treating these diseases in clinical practice are continuously evolving and have rapidly revolutionized treatment opinions. However, key issues such as inadequate treatment effectiveness, high rates of recurrence, and poor patient compliance still need to be urgently addressed. Multifunctional nanomedicine can specifically respond to both endogenous and exogenous microenvironments, effectively deliver drugs to specific targets and participate in activities such as biological imaging and the detection of small molecules. Nano-in-micro (NIM) delivery systems such as metal, metal oxide and up-conversion nanoparticles (NPs), quantum dots, and carbon materials, have shown certain advantages in overcoming the presence of physiological barriers within the eyeball and are widely used in the treatment of ophthalmic diseases. Few studies, however, have evaluated the efficacy of NIM delivery systems in treating fundus neovascular diseases (FNDs). The present study describes the main clinical treatment strategies and the adverse events associated with the treatment of FNDs with NIM delivery systems and summarizes the anatomical obstacles that must be overcome. In this review, we wish to highlight the principle of intraocular microenvironment normalization, aiming to provide a more rational approach for designing new NIM delivery systems to treat specific FNDs.


Asunto(s)
Sistemas de Liberación de Medicamentos , Humanos , Animales , Sistemas de Liberación de Medicamentos/métodos , Neovascularización Patológica/tratamiento farmacológico , Fondo de Ojo , Puntos Cuánticos/química , Nanopartículas Multifuncionales/química , Neovascularización Retiniana/tratamiento farmacológico , Nanomedicina/métodos , Nanopartículas/química
8.
Biol Res ; 57(1): 43, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38915069

RESUMEN

BACKGROUND: Retinopathy of Prematurity (ROP) is a proliferative retinal vascular disease occurring in the retina of premature infants and is the main cause of childhood blindness. Nowadays anti-VEGF and retinal photocoagulation are mainstream treatments for ROP, but they develop a variety of complications. Hydrogen (H2) is widely considered as a useful neuroprotective and antioxidative therapeutic method for hypoxic-ischemic disease without toxic effects. However, whether H2 provides physiological angiogenesis promotion, neovascularization suppression and glial protection in the progression of ROP is largely unknown.This study aims to investigate the effects of H2 on retinal angiogenesis, neovascularization and neuroglial dysfunction in the retinas of oxygen-induced retinopathy (OIR) mice. METHODS: In this study, mice that were seven days old and either wild-type (WT) or Nrf2-deficient (Nrf2-/-) were exposed to 75% oxygen for 5 days and then returned to normal air conditions. Different stages of hydrogen gas (H2) inhalation were administered. Vascular obliteration, neovascularization, and blood vessel leakage were analyzed and compared. To count the number of neovascularization endothelial nuclei, routine HE staining of retinal sections was conducted. Immunohistochemistry was performed using DyLight 594 labeled GSL I-isolectin B4 (IB4), as well as primary antibodies against proliferating cell nuclear antigen (PCNA), glial fibrillary acidic protein (GFAP), and Iba-1. Western blots were used to measure the expression of NF-E2-related factor 2 (Nrf2), vascular endothelial growth factor (VEGF), Notch1, Dll4, and HIF-1α. Additionally, the expression of target genes such as NQO1, HO-1, Notch1, Hey1, Hey2, and Dll4 was measured. Human umbilical vein endothelial cells (HUVECs) treated with H2 under hypoxia were used as an in vitro model. RT-PCR was used to evaluate the mRNA expression of Nrf2, Notch/Dll4, and the target genes. The expression of reactive oxygen species (ROS) was observed using immunofluorescence staining. RESULTS: Our results indicate that 3-4% H2 does not disturb retinal physiological angiogenesis, but ameliorates vaso-obliteration and neovascularization in OIR mice. Moreover, H2 prevents the decreased density and reverses the morphologic and functional changes in retinal astrocytes caused by oxygen-induced injury. In addition, H2 inhalation reduces microglial activation, especially in the area of neovascularization in OIR mice. H2 plays a protective role in vascular regeneration by promoting Nrf2 activation and suppressing the Dll4-induced Notch signaling pathway in vivo. Also, H2 promotes the proliferation of HUVECs under hypoxia by negatively regulating the Dll4/Notch pathway and reducing ROS levels through Nrf2 pathway aligning with our findings in vivo.Moreover, the retinal oxygen-sensing mechanisms (HIF-1α/VEGF) are also involved in hydrogen-mediated retinal revascularization and neovascularization suppression. CONCLUSIONS: Collectively, our results indicate that H2 could be a promising therapeutic agent for POR treatment and that its beneficial effect in human ROP might involve the activation of the Nrf2-Notch axis as well as HIF-1α/VEGF pathways.


Asunto(s)
Modelos Animales de Enfermedad , Hidrógeno , Neuroglía , Oxígeno , Neovascularización Retiniana , Retinopatía de la Prematuridad , Animales , Hidrógeno/farmacología , Neovascularización Retiniana/tratamiento farmacológico , Neuroglía/efectos de los fármacos , Ratones , Retinopatía de la Prematuridad/tratamiento farmacológico , Ratones Endogámicos C57BL , Retina/efectos de los fármacos , Animales Recién Nacidos , Regeneración/efectos de los fármacos , Inmunohistoquímica , Vasos Retinianos/efectos de los fármacos
9.
Sci Rep ; 14(1): 13399, 2024 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-38862630

RESUMEN

This retrospective observational study aimed to investigate the difference in 4-year outcomes of ranibizumab or aflibercept therapy for macular neovascularization (MNV) with high myopia between pathologic myopia (PM) and non-PM. This study was conducted at Kyoto University Hospital and included consecutive treatment-naïve eyes with active myopic MNV, in which a single intravitreal ranibizumab or aflibercept injection was administered, followed by a pro re nata (PRN) regimen for 4 years. Based on the META-PM study classification, eyes were assigned to the non-PM and PM groups. This study analyzed 118 eyes of 118 patients (non-PM group, 19 eyes; PM group, 99 eyes). Baseline, 1-year, and 2-year best-corrected visual acuity (BCVA) were significantly better in the non-PM group (P = 0.02, 0.01, and 0.02, respectively); however, the 3-year and 4-year BCVA were not. The 4-year BCVA course was similar in both groups. However, the total number of injections over 4 years was significantly higher in the non-PM than in the PM group (4.6 ± 2.6 vs. 2.9 ± 2.6, P = 0.001). Four-year BCVA significantly correlated only with baseline BCVA in both non-PM (P = 0.047, ß = 0.46) and PM groups (P < 0.001, ß = 0.59). In conclusion, over the 4-year observation period, the BCVA course after anti-VEGF therapy for myopic MNV was similar in the eyes with non-PM and those with PM; however, more additional injections in a PRN regimen were required in the eyes with non-PM compared to those with PM. Thus, more frequent and careful follow-up is required for the eyes with non-PM compared with those with PM to maintain long-term BCVA.


Asunto(s)
Inhibidores de la Angiogénesis , Miopía Degenerativa , Ranibizumab , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión , Factor A de Crecimiento Endotelial Vascular , Agudeza Visual , Humanos , Masculino , Femenino , Ranibizumab/administración & dosificación , Ranibizumab/uso terapéutico , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/uso terapéutico , Receptores de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Anciano , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Estudios Retrospectivos , Resultado del Tratamiento , Persona de Mediana Edad , Miopía Degenerativa/tratamiento farmacológico , Miopía Degenerativa/complicaciones , Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/administración & dosificación , Inyecciones Intravítreas , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/patología , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/patología
10.
Angiogenesis ; 27(3): 423-440, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38709389

RESUMEN

BACKGROUND: Retinopathy of prematurity (ROP), which often presents with bronchopulmonary dysplasia (BPD), is among the most common morbidities affecting extremely premature infants and is a leading cause of severe vision impairment in children worldwide. Activations of the inflammasome cascade and microglia have been implicated in playing a role in the development of both ROP and BPD. Apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) is pivotal in inflammasome assembly. Utilizing mouse models of both oxygen-induced retinopathy (OIR) and BPD, this study was designed to test the hypothesis that hyperoxia induces ASC speck formation, which leads to microglial activation and retinopathy, and that inhibition of ASC speck formation by a humanized monoclonal antibody, IC100, directed against ASC, will ameliorate microglial activation and abnormal retinal vascular formation. METHODS: We first tested ASC speck formation in the retina of ASC-citrine reporter mice expressing ASC fusion protein with a C-terminal citrine (fluorescent GFP isoform) using a BPD model that causes both lung and eye injury by exposing newborn mice to room air (RA) or 85% O2 from postnatal day (P) 1 to P14. The retinas were dissected on P14 and retinal flat mounts were used to detect vascular endothelium with AF-594-conjugated isolectin B4 (IB4) and citrine-tagged ASC specks. To assess the effects of IC100 on an OIR model, newborn ASC citrine reporter mice and wildtype mice (C57BL/6 J) were exposed to RA from P1 to P6, then 75% O2 from P7 to P11, and then to RA from P12 to P18. At P12 mice were randomized to the following groups: RA with placebo PBS (RA-PBS), O2 with PBS (O2-PBS), O2 + IC100 intravitreal injection (O2-IC100-IVT), and O2 + IC100 intraperitoneal injection (O2-IC100-IP). Retinal vascularization was evaluated by flat mount staining with IB4. Microglial activation was detected by immunofluorescence staining for allograft inflammatory factor 1 (AIF-1) and CD206. Retinal structure was analyzed on H&E-stained sections, and function was analyzed by pattern electroretinography (PERG). RNA-sequencing (RNA-seq) of the retinas was performed to determine the transcriptional effects of IC100 treatment in OIR. RESULTS: ASC specks were significantly increased in the retinas by hyperoxia exposure and colocalized with the abnormal vasculature in both BPD and OIR models, and this was associated with increased microglial activation. Treatment with IC100-IVT or IC100-IP significantly reduced vaso-obliteration and intravitreal neovascularization. IC100-IVT treatment also reduced retinal microglial activation, restored retinal structure, and improved retinal function. RNA-seq showed that IC100 treatment corrected the induction of genes associated with angiogenesis, leukocyte migration, and VEGF signaling caused by O2. IC100 also corrected the suppression of genes associated with cell junction assembly, neuron projection, and neuron recognition caused by O2. CONCLUSION: These data demonstrate the crucial role of ASC in the pathogenesis of OIR and the efficacy of a humanized therapeutic anti-ASC antibody in treating OIR mice. Thus, this anti-ASC antibody may potentially be considered in diseases associated with oxygen stresses and retinopathy, such as ROP.


Asunto(s)
Oxígeno , Retinopatía de la Prematuridad , Animales , Retinopatía de la Prematuridad/patología , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/metabolismo , Ratones , Anticuerpos Monoclonales Humanizados/farmacología , Ratones Endogámicos C57BL , Animales Recién Nacidos , Modelos Animales de Enfermedad , Humanos , Hiperoxia/patología , Hiperoxia/complicaciones , Retina/patología , Retina/metabolismo , Retina/efectos de los fármacos , Proteínas Adaptadoras de Señalización CARD/metabolismo , Ratones Transgénicos , Neovascularización Retiniana/patología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/tratamiento farmacológico , Microglía/patología , Microglía/metabolismo , Microglía/efectos de los fármacos
11.
Immunology ; 173(1): 141-151, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38804253

RESUMEN

Retinopathy of prematurity (ROP) is a retinal disease-causing retinal neovascularization that can lead to blindness. Oxygen-induced retinopathy (OIR) is a widely used ROP animal model. Icariin (ICA) has anti-oxidative and anti-inflammation properties; however, whether ICA has a regulatory effect on OIR remains unclear. In this study, ICA alleviated pathological neovascularization, microglial activation and blood-retina barrier (BRB) damage in vivo. Further results indicated that endothelial cell tube formation, migration and proliferation were restored by ICA treatment in vitro. Proteomic microarrays and molecular mimicry revealed that ICA can directly bind to hexokinase 2 (HK2) and decrease HK2 protein expression in vivo and in vitro. In addition, ICA inhibited the AKT/mTOR/HIF1α pathway activation. The effects of ICA on pathological neovascularization, microglial activation and BRB damage disappeared after HK2 overexpression in vivo. Similarly, the endothelial cell function was revised after HK2 overexpression. HK2 overexpression reversed ICA-induced AKT/mTOR/HIF1α pathway inhibition in vivo and in vitro. Therefore, ICA prevented pathological angiogenesis in OIR in an HK2-dependent manner, implicating ICA as a potential therapeutic agent for ROP.


Asunto(s)
Flavonoides , Hexoquinasa , Microglía , Oxígeno , Neovascularización Retiniana , Retinopatía de la Prematuridad , Transducción de Señal , Serina-Treonina Quinasas TOR , Animales , Humanos , Ratones , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Flavonoides/farmacología , Flavonoides/uso terapéutico , Hexoquinasa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/metabolismo , Retinopatía de la Prematuridad/patología , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo
12.
Biochem Biophys Res Commun ; 720: 150065, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38749188

RESUMEN

Neovascular eye diseases, including proliferative diabetic retinopathy and retinopathy of prematurity, is a major cause of blindness. Laser ablation and intravitreal anti-VEGF injection have shown their limitations in treatment of retinal neovascularization. Identification of a new therapeutic strategies is in urgent need. Our study aims to assess the effects of Cryptotanshinone (CPT), a natural compound derived from Salvia miltiorrhiza Bunge, in retina neovascularization and explore its potential mechanism. Our study demonstrated that CPT did not cause retina tissue toxicity at the tested concentrations. Intravitreal injections of CPT reduced pathological angiogenesis and promoted physical angiogenesis in oxygen-induced retinopathy (OIR) model. CPT improve visual function in OIR mice and reduced cell apoptosis. Moreover, we also revealed that CPT diminishes the expression of inflammatory cytokines in the OIR retina. In vitro, the administration of CPT effectively inhibited endothelial cells proliferation, migration, sprouting, and tube formation induced by the stimulation of human retinal vascular endothelial cells (HRVECs) with VEGF165. Mechanistically, CPT blocking the phosphorylation of VEGFR2 and downstream targeting pathway. After all, the findings demonstrated that CPT exhibits potent anti-angiogenic and anti-inflammatory effects in OIR mice, and it has therapeutic potential for the treatment of neovascular retinal diseases.


Asunto(s)
Inyecciones Intravítreas , Ratones Endogámicos C57BL , Fenantrenos , Neovascularización Retiniana , Animales , Fenantrenos/farmacología , Fenantrenos/administración & dosificación , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/patología , Neovascularización Retiniana/metabolismo , Humanos , Ratones , Inhibidores de la Angiogénesis/farmacología , Inhibidores de la Angiogénesis/administración & dosificación , Proliferación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Retina/efectos de los fármacos , Retina/metabolismo , Retina/patología
13.
BMC Ophthalmol ; 24(1): 224, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38807066

RESUMEN

BACKGROUND: Macular retinoschisis (MRS) and myopic macular neovascularization (mMNV) are both potentially blinding complications of high myopia. In this case report, we highlight the progression of MRS after intravitreal anti-vascular endothelial growth factor (anti-VEGF) treatment for mMNV, as well as an extensive review of the literature on this topic. CASE DESCRIPTION: A 49-year-old woman presented with two weeks of recent onset blurring and metamorphopsia in her right eye. She had high myopia in both eyes (right eye - 20/60 with - 16D, left eye - 20/20 with - 13D). Slit-lamp ophthalmoscopy found a normal anterior segment in both eyes. On fundus examination, features of pathological myopia with posterior staphyloma and peripapillary atrophy were observed in both eyes. An active mMNV, as well as intraretinal fluid, minimal perifoveal inner and outer MRS, and focal posterior vitreous traction along the inferotemporal retinal arcade, were detected on optical coherence tomography (OCT) of the right eye. The patient received an intravitreal injection of Aflibercept (2 mg/0.05 ml). RESULTS: OCT scans at two- and four-month follow-up visits revealed regressed mMNV with a taut epiretinal membrane, progressive worsening of outer MRS, and the development of multiple perifoveal retinal detachment inferior to the fovea. Pars plana vitrectomy surgery was performed for the progressive MRS with good anatomical (resolved MRS) and functional outcome (maintained visual acuity at 20/60) at the last one-month post-surgery visit. CONCLUSION: Intravitreal anti-VEGF injections for mMNV can cause vitreoretinal interface changes, exacerbating MRS and causing visual deterioration. Vitrectomy for MRS could be one of several treatment options.


Asunto(s)
Inyecciones Intravítreas , Miopía Degenerativa , Receptores de Factores de Crecimiento Endotelial Vascular , Proteínas Recombinantes de Fusión , Retinosquisis , Tomografía de Coherencia Óptica , Agudeza Visual , Humanos , Receptores de Factores de Crecimiento Endotelial Vascular/uso terapéutico , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/administración & dosificación , Femenino , Persona de Mediana Edad , Retinosquisis/diagnóstico , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Miopía Degenerativa/complicaciones , Inhibidores de la Angiogénesis/efectos adversos , Inhibidores de la Angiogénesis/administración & dosificación , Progresión de la Enfermedad , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/diagnóstico , Neovascularización Retiniana/inducido químicamente , Angiografía con Fluoresceína
14.
BMC Pediatr ; 24(1): 301, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704544

RESUMEN

BACKGROUND: We present a patient with retinopathy of prematurity (ROP) who developed worsening plus disease after complete regression of stage 3 ROP. The use of fundus fluorescein angiography (FFA) aided the visualization of occult neovascularization that caused the disease progression. CASE PRESENTATION: The patient was at high risk for ROP due to low birth weight of 690 g and gestational age of 25 weeks. After the diagnosis of stage 3 ROP in zone I without plus disease, she was treated initially with bilateral intravitreal bevacizumab (IVB) and followed by laser photocoagulation 5 weeks later. Despite the resolution of ROP stage, the plus disease worsened. Neither systemic risk factors nor skip laser areas were observed. Hence, FFA was performed and subsequently identified occult neovascularization with active leakage. Additional IVB and laser treatment in the capillary dropout area inside vascularized retina were added. The plus disease improved but mild arteriolar tortuosity persisted. CONCLUSIONS: Worsening of plus disease after completion of laser ablation and IVB with complete regression of stage 3 ROP is rare. Systemic risk factors such as continuous oxygen therapy and cardiovascular disease should be ruled out. FFA aided in identifying occult neovascularization and prompted further treatment.


Asunto(s)
Inhibidores de la Angiogénesis , Bevacizumab , Angiografía con Fluoresceína , Inyecciones Intravítreas , Coagulación con Láser , Neovascularización Retiniana , Retinopatía de la Prematuridad , Humanos , Bevacizumab/uso terapéutico , Bevacizumab/administración & dosificación , Recién Nacido , Inhibidores de la Angiogénesis/uso terapéutico , Inhibidores de la Angiogénesis/administración & dosificación , Femenino , Coagulación con Láser/métodos , Neovascularización Retiniana/etiología , Neovascularización Retiniana/tratamiento farmacológico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/administración & dosificación , Terapia Combinada
15.
Acta Pharmacol Sin ; 45(7): 1438-1450, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38565961

RESUMEN

Angiogenesis plays a critical role in many pathological processes, including irreversible blindness in eye diseases such as retinopathy of prematurity. Endothelial mitochondria are dynamic organelles that undergo constant fusion and fission and are critical signalling hubs that modulate angiogenesis by coordinating reactive oxygen species (ROS) production and calcium signalling and metabolism. In this study, we investigated the role of mitochondrial dynamics in pathological retinal angiogenesis. We showed that treatment with vascular endothelial growth factor (VEGF; 20 ng/ml) induced mitochondrial fission in HUVECs by promoting the phosphorylation of dynamin-related protein 1 (DRP1). DRP1 knockdown or pretreatment with the DRP1 inhibitor Mdivi-1 (5 µM) blocked VEGF-induced cell migration, proliferation, and tube formation in HUVECs. We demonstrated that VEGF treatment increased mitochondrial ROS production in HUVECs, which was necessary for HIF-1α-dependent glycolysis, as well as proliferation, migration, and tube formation, and the inhibition of mitochondrial fission prevented VEGF-induced mitochondrial ROS production. In an oxygen-induced retinopathy (OIR) mouse model, we found that active DRP1 was highly expressed in endothelial cells in neovascular tufts. The administration of Mdivi-1 (10 mg·kg-1·d-1, i.p.) for three days from postnatal day (P) 13 until P15 significantly alleviated pathological angiogenesis in the retina. Our results suggest that targeting mitochondrial fission may be a therapeutic strategy for proliferative retinopathies and other diseases that are dependent on pathological angiogenesis.


Asunto(s)
Movimiento Celular , Dinaminas , Células Endoteliales de la Vena Umbilical Humana , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones Endogámicos C57BL , Dinámicas Mitocondriales , Quinazolinonas , Especies Reactivas de Oxígeno , Neovascularización Retiniana , Factor A de Crecimiento Endotelial Vascular , Dinámicas Mitocondriales/efectos de los fármacos , Animales , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Dinaminas/metabolismo , Dinaminas/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Quinazolinonas/farmacología , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Neovascularización Retiniana/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Ratones , Proliferación Celular/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Angiogénesis
16.
Eur J Pharm Biopharm ; 199: 114296, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38636882

RESUMEN

Small interfering RNA (siRNA) is emerging as a promising treatment for retinal neovascularization due to its specific inhibition of the expression of target genes. However, the clinical translation of siRNA drugs is hindered by the efficiency and safety of delivery vectors. Here, we describe the properties of a new bioreducible ionizable lipid nanoparticle (LNP) 2N12H, which is based on a rationally designed novel ionizable lipid called 2N12B. 2N12H exhibited degradation in response to the mimic cytoplasmic glutathione condition and ionization with a pKa value of 6.5, which remaining neutral at pH 7.4. At a nitrogen to phosphorus ratio of 5, 2N12H efficiently encapsulated and protected siRNA from degradation. Compared to the commercial vehicle Lipofectamine 2000, 2N12H demonstrated similar silencing efficiency and improved safety in the in vitro cell experiments. 2N12H/siVEGFA reduced the expression of VEGFA in retinal pigment epithelium cells and mouse retina, consequently suppressing cell migration and retinal neovascularization. In the mouse model, the therapeutic effect of 2N12H/siVEGFA was comparable to that of the clinical drug ranibizumab. Together, these results suggest the potential of this novel ionizable LNP to facilitate the development of nonviral ocular gene delivery systems.


Asunto(s)
Lípidos , Ratones Endogámicos C57BL , Nanopartículas , ARN Interferente Pequeño , Neovascularización Retiniana , Factor A de Crecimiento Endotelial Vascular , Animales , Nanopartículas/química , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/farmacología , Neovascularización Retiniana/tratamiento farmacológico , Ratones , Lípidos/química , Humanos , Factor A de Crecimiento Endotelial Vascular/genética , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Movimiento Celular/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Ranibizumab/administración & dosificación , Técnicas de Transferencia de Gen , Retina/metabolismo , Retina/efectos de los fármacos
17.
Invest Ophthalmol Vis Sci ; 65(4): 37, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38652648

RESUMEN

Purpose: Adjuvant, pre-operative intravitreal anti-vascular endothelial growth factor (anti-VEGF) injections have been used to reduce peri-operative bleeding in eyes undergoing pars-plana vitrectomy for complications of proliferative diabetic retinopathy (PDR). To address the concern over their potential off-target effects of progressive fibrous contraction, we sought to dissect the transcriptional changes in the surgically extracted fibrovascular membranes (FVMs). Methods: We analyzed surgically extracted FVMs from 10 eyes: 4 eyes pretreated with intravitreal bevacizumab (IVB) and 6 untreated eyes. FVMs were digested into single cells, mRNA was extracted from endothelial cell-enriched (microbead selection with CD31) and non-endothelial cell compartments, followed by RT-qPCR quantification. We then compared the relative expression of genes involved in angiogenesis, endothelial cell integrity, and myofibroblastic processes between treated and untreated FVMs. Results: Endothelial cells from IVB pretreated FVMs showed significant reduction of VEGFA, VEGF receptors (FLT1 and KDR), and angiopoietin 2 expression as well as increased vascular endothelial cadherin and endothelin, suggesting reduced angiogenesis and enhanced vascular integrity. The non-endothelial cell fraction showed decreased expression of VEGFA and fibronectin, without significant difference in the expression of other profibrotic factors. Conclusions: Our findings confirm that adjuvant pre-operative IVB decreased fibronectin and increase endothelin-1 expression without affecting other profibrotic gene expression, uncovering an important interaction between IVB and endothelin-1 that deserves further study.


Asunto(s)
Inhibidores de la Angiogénesis , Bevacizumab , Retinopatía Diabética , Fibrosis , Inyecciones Intravítreas , Factor A de Crecimiento Endotelial Vascular , Vitrectomía , Humanos , Retinopatía Diabética/metabolismo , Retinopatía Diabética/tratamiento farmacológico , Retinopatía Diabética/cirugía , Inhibidores de la Angiogénesis/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Bevacizumab/uso terapéutico , Bevacizumab/farmacología , Masculino , Femenino , Persona de Mediana Edad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/tratamiento farmacológico , Anciano , Cuidados Preoperatorios , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/farmacología
18.
Ocul Immunol Inflamm ; 32(5): 529-533, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38436937

RESUMEN

PURPOSE: To report a rare case of cytomegalovirus (CMV)-associated non-necrotizing viral retinopathy, occlusive retinal vasculitis, papillitis, and retinal neovascularization in a young 41-year-old woman. METHODS: Case report. RESULTS: The patient presented with features of papillitis, peripapillary cotton-wool spots, pre-retinal hemorrhages, and occlusive vasculitis. Her visual acuity was 20/100 in the left eye. She developed a worsening of the disease upon initiation of systemic corticosteroids. Her serum immunoglobulins (Ig) (both IgG and IgM) were highly positive for CMV. Anterior chamber paracentesis was positive for CMV DNA using real-time polymerase chain reaction. After stopping systemic corticosteroids, she was initiated on oral valganciclovir, with rapid resolution of the vasculitis and cotton-wool spots. After three months, the patient developed retinal neovascularization and underwent pan-retinal photocoagulation. However, her uveitis was inactive, and her visual acuity improved to 20/25. CONCLUSIONS: Non-necrotizing viral retinopathy has been associated with either varicella zoster virus (VZV) or herpes simplex virus (HSV). Our case highlights that CMV can also lead to non-necrotizing retinopathy and must be suspected in patients who may be negative for VZV and HSV. Appropriate anti-viral treatment can prevent severe vision loss in these patients.


Asunto(s)
Antivirales , Citomegalovirus , ADN Viral , Infecciones Virales del Ojo , Angiografía con Fluoresceína , Neovascularización Retiniana , Vasculitis Retiniana , Agudeza Visual , Humanos , Femenino , Adulto , Vasculitis Retiniana/diagnóstico , Vasculitis Retiniana/virología , Vasculitis Retiniana/tratamiento farmacológico , Neovascularización Retiniana/diagnóstico , Neovascularización Retiniana/tratamiento farmacológico , Neovascularización Retiniana/etiología , Neovascularización Retiniana/virología , Citomegalovirus/genética , Citomegalovirus/aislamiento & purificación , Infecciones Virales del Ojo/diagnóstico , Infecciones Virales del Ojo/virología , Infecciones Virales del Ojo/tratamiento farmacológico , Antivirales/uso terapéutico , ADN Viral/análisis , Retinitis por Citomegalovirus/diagnóstico , Retinitis por Citomegalovirus/tratamiento farmacológico , Tomografía de Coherencia Óptica , Valganciclovir/uso terapéutico , Fondo de Ojo
19.
Cell Prolif ; 57(7): e13623, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38433462

RESUMEN

The objective of this study was to investigate the effects and molecular mechanisms of tetrahedral framework nucleic acids-microRNA22 (tFNAs-miR22) on inhibiting pathological retinal neovascularization (RNV) and restoring physiological retinal vessels. A novel DNA nanocomplex (tFNAs-miR22) was synthesised by modifying microRNA-22 (miR22) through attachment onto tetrahedral frame nucleic acids (tFNAs), which possess diverse biological functions. Cell proliferation, wound healing, and tube formation were employed for in vitro assays to investigate the angiogenic function of cells. Oxygen-induced retinopathy (OIR) model was utilised to examine the effects of reducing pathological neovascularization (RNV) and inhibiting vascular occlusion in vivo. In vitro, tFNAs-miR22 demonstrated the ability to penetrate endothelial cells and effectively suppress cell proliferation, tube formation, and migration in a hypoxic environment. In vivo, tFNAs-miR22 exhibited promising results in reducing RNV and promoting the restoration of normal retinal blood vessels in OIR model through modulation of the Wnt pathway. This study provided a theoretical basis for the further understanding of RNV, and highlighted the innovative and potential of tFNAs-miR22 as a therapeutic option for ischemic retinal diseases.


Asunto(s)
Proliferación Celular , MicroARNs , Neovascularización Retiniana , Vía de Señalización Wnt , MicroARNs/genética , MicroARNs/metabolismo , Animales , Vía de Señalización Wnt/efectos de los fármacos , Humanos , Proliferación Celular/efectos de los fármacos , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología , Neovascularización Retiniana/tratamiento farmacológico , Ratones , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Movimiento Celular/efectos de los fármacos , Neovascularización Patológica/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Ratones Endogámicos C57BL , Ácidos Nucleicos/metabolismo
20.
Int Ophthalmol ; 44(1): 134, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38483688

RESUMEN

PURPOSE: The aim of this study was to compare the responses of type 1 and type 2 macular neovascularizations (MNV) caused by neovascular type age-related macular degeneration (n-AMD) to intravitreal anti-vascular endothelial growth factor (VEGF) treatments using quantitative parameters determined by optical coherence tomography (OCT). Additionally, it was also intended to assess the connections between these quantitative parameters and changes in best-corrected visual acuity (BCVA) and the number of intravitreal anti-VEGF injections required within a year. MATERIALS AND METHODS: In our retrospective and observational study, the data of 90 eyes of 90 patients diagnosed with n-AMD and treated with intravitreal anti-VEGF with the "Pro re nata" method were evaluated. Subtypes of existing MNVs were distinguished with previously taken optical coherence tomography angiography (OCTA) images. In spectral domain OCT examinations, central macular thickness (CMT) and central macular volume (CMV) values were recorded at baseline and 12th month. The number of intravitreal anti-VEGF injections during the 12 month follow-up period was also recorded for each patient. Obtained data were compared between MNV types. RESULTS: Of the n-AMD cases examined in the study, 56.66% had type 1 MNV and 43.34% had type 2 MNV. The mean baseline BCVA logMAR values in eyes with type 2 MNV (1.15 ± 0.43) were higher than those observed in eyes with type 1 MNV (0.76 ± 0.42) (p = 0.001). Similarly, mean baseline CMT and CMV values in eyes with type 2 MNV were higher than those observed in eyes with type 1 MNV (respectively 424.89 ± 49.46 µm vs. 341.39 ± 37.06 µm; 9.17 ± 0.89 µm3 vs. 8.49 ± 0.53 µm3; p < 0.05). After 12 months of treatment, logMAR values of BCVA (0.86 ± 0.42) in subjects with type 2 MNV were higher than those in subjects with type 1 MNV (0.57 ± 0.37) (p = 0.001). Mean CMT and CMV values at 12th month in subjects with type 2 MNV (379.11 ± 46.36 µm and 8.66 ± 0.79 µm3, respectively) were observed to be higher than those with type 1 MNV (296.95 ± 33.96 µm and 8.01 ± 0.52 mm3, respectively) (p < 0.05). In type 2 MNVs, positive correlations were observed between both baseline and 12th month BCVA logMAR values and baseline CMV (p < 0.05). Similarly, in type 2 MNVs, a positive correlation was observed between 12th month BCVA logMAR values and 12th month CMV (p < 0.05). The total number of intravitreal anti-VEGF injections at 12 months was similar in both groups (p = 0.851). CONCLUSION: In this study, in which we performed a subtype analysis of MNV cases, we observed that the visual function was worse at the beginning and the end of the 12th month, and the CMT and CMV values were higher in the type 2 MNV group compared to the type 1 MNV cases. In addition, we found significant correlations between BCVA logMAR values and CMV values in type 2 MNV cases. In the follow-up of these cases, CMT, which is a more widely used quantitative method, and CMV, which is a newer OCT measurement parameter, may be more useful in patient follow-up and evaluation of treatment efficacy, especially for type 2 MNV cases.


Asunto(s)
Infecciones por Citomegalovirus , Degeneración Macular , Neovascularización Retiniana , Humanos , Inhibidores de la Angiogénesis , Tomografía de Coherencia Óptica/métodos , Estudios de Seguimiento , Estudios Retrospectivos , Angiografía con Fluoresceína/métodos , Neovascularización Retiniana/tratamiento farmacológico , Inyecciones Intravítreas , Degeneración Macular/complicaciones , Infecciones por Citomegalovirus/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA