Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 282
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Angiogenesis ; 27(3): 311-331, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38564108

RESUMO

Diabetic retinopathy has a high probability of causing visual impairment or blindness throughout the disease progression and is characterized by the growth of new blood vessels in the retina at an advanced, proliferative stage. Microglia are a resident immune population in the central nervous system, known to play a crucial role in regulating retinal angiogenesis in both physiological and pathological conditions, including diabetic retinopathy. Physiologically, they are located close to blood vessels and are essential for forming new blood vessels (neovascularization). In diabetic retinopathy, microglia become widely activated, showing a distinct polarization phenotype that leads to their accumulation around neovascular tufts. These activated microglia induce pathogenic angiogenesis through the secretion of various angiogenic factors and by regulating the status of endothelial cells. Interestingly, some subtypes of microglia simultaneously promote the regression of neovascularization tufts and normal angiogenesis in neovascularization lesions. Modulating the state of microglial activation to ameliorate neovascularization thus appears as a promising potential therapeutic approach for managing diabetic retinopathy.


Assuntos
Retinopatia Diabética , Microglia , Neovascularização Retiniana , Animais , Humanos , Angiogênese/metabolismo , Angiogênese/patologia , Retinopatia Diabética/patologia , Retinopatia Diabética/metabolismo , Microglia/patologia , Microglia/metabolismo , Retina/patologia , Retina/metabolismo , Neovascularização Retiniana/patologia , Neovascularização Retiniana/metabolismo , Vasos Retinianos/patologia , Vasos Retinianos/metabolismo
2.
Microvasc Res ; 154: 104689, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38636926

RESUMO

Pathological retinal angiogenesis is not only the hallmark of retinopathies, but also a major cause of blindness. Guanylate binding protein 2 (GBP2) has been reported to be associated with retinal diseases such as diabetic retinopathy and hypoxic retinopathy. However, GBP2-mediated pathological retinal angiogenesis remains largely unknown. The present study aimed to investigate the role of GBP2 in pathological retinal angiogenesis and its underlying molecular mechanism. In this study, we established oxygen-induced retinopathy (OIR) mice model for in vivo study and hypoxia-induced angiogenesis in ARPE-19 cells for in vitro study. We demonstrated that GBP2 expression was markedly downregulated in the retina of mice with OIR and ARPE-19 cells treated with hypoxia, which was associated with pathological retinal angiogenesis. The regulatory mechanism of GBP2 in ARPE-19 cells was studied by GBP2 silencing and overexpression. The regulatory mechanism of GBP2 in the retina was investigated by overexpressing GBP2 in the retina of OIR mice. Mechanistically, GBP2 downregulated the expression and secretion of vascular endothelial growth factor (VEGFA) in ARPE-19 cells and retina of OIR mice. Interestingly, overexpression of GBP2 significantly inhibited neovascularization in OIR mice, conditioned medium of GBP2 overexpressing ARPE-19 cells inhibited angiogenesis in human umbilical vein endothelial cells (HUVECs). Furthermore, we confirmed that GBP2 downregulated VEGFA expression and angiogenesis by inhibiting the AKT/mTOR signaling pathway. Taken together, we concluded that GBP2 inhibited pathological retinal angiogenesis via the AKT/mTOR/VEGFA axis, thereby suggesting that GBP2 may be a therapeutic target for pathological retinal angiogenesis.


Assuntos
Modelos Animais de Doenças , Proteínas de Ligação ao GTP , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-akt , Neovascularização Retiniana , Vasos Retinianos , Transdução de Sinais , Serina-Treonina Quinases TOR , Fator A de Crescimento do Endotélio Vascular , Animais , Humanos , Camundongos , Hipóxia Celular , Linhagem Celular , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/genética , Oxigênio/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/prevenção & controle , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Serina-Treonina Quinases TOR/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética
3.
Exp Eye Res ; 248: 110072, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39241859

RESUMO

The study aims to investigate the effects and potential mechanisms of lncRNA-MM2P on retinal neovascularization in a mouse model of oxygen-induced retinopathy (OIR). The OIR model was established in C57BL/6J mice. RAW264.7 cell line and bone marrow-derived macrophages (BMDMs) from mice were used for in vitro studies. RT-qPCR was used to analyze the expressions of lncRNA and mRNAs. The protein expression levels were determined by western blotting. The size of avascular areas and neovascular tufts were assessed based on isolectin B4 immunofluorescence staining images. The human retinal endothelial cells (HRECs) were used to evaluate the proliferation, migration, and tube formation of endothelial cells. The expression of lncRNA-MM2P was significantly upregulated from P17 to P25 in OIR retinas. Knockdown of lncRNA-MM2P levels in vivo led to a significant reduction in the neovascular tufts and avascular areas in the retinas of OIR mice. Knockdown of lncRNA-MM2P levels in vitro suppressed the expression of M2 markers in macrophages. Moreover, we found a significant inhibition of avascular areas and neovascular tufts in OIR mice injected intravitreally with M2 macrophages treated by shRNA-MM2P. The cellular functions of proliferation, migration, and tube formation were significantly attenuated in HRECs cultured with a supernatant of shRNA-MM2P-treated M2 macrophages. Our results indicate that lncRNA-MM2P regulates retinal neovascularization by inducing M2 polarization of macrophages in OIR mice. Therefore, lncRNA-MM2P may be a potential molecular target for immunoregulation of retinal neovascularization.

4.
Exp Eye Res ; 242: 109870, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38514023

RESUMO

Retinal neovascularization (RNV) is a pathological process that primarily occurs in diabetic retinopathy, retinopathy of prematurity, and retinal vein occlusion. It is a common yet debilitating clinical condition that culminates in blindness. Urgent efforts are required to explore more efficient and less limiting therapeutic strategies. Key RNA-binding proteins (RBPs), crucial for post-transcriptional regulation of gene expression by binding to RNAs, are closely correlated with RNV development. RBP-RNA interactions are altered during RNV. Here, we briefly review the characteristics and functions of RBPs, and the mechanism of RNV. Then, we present insights into the role of the regulatory network of RBPs in RNV. HuR, eIF4E, LIN28B, SRSF1, METTL3, YTHDF1, Gal-1, HIWI1, and ZFR accelerate RNV progression, whereas YTHDF2 and hnRNPA2B1 hinder it. The mechanisms elucidated in this review provide a reference to guide the design of therapeutic strategies to reverse abnormal processes.


Assuntos
Proteínas de Ligação a RNA , Neovascularização Retiniana , Humanos , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação da Expressão Gênica/fisiologia , Animais
5.
Arterioscler Thromb Vasc Biol ; 43(4): 522-536, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36794587

RESUMO

BACKGROUND: CD4+ (cluster of differentation) and CD8+ T cells are increased in the ocular fluids of patients with neovascular retinopathy, yet their role in the disease process is unknown. METHODS: We describe how CD8+ T cells migrate into the retina and contribute to pathological angiogenesis by releasing cytokines and cytotoxic factors. RESULTS: In oxygen-induced retinopathy, flow cytometry revealed the numbers of CD4+ and CD8+ T cells were increased in blood, lymphoid organs, and retina throughout the development of neovascular retinopathy. Interestingly, the depletion of CD8+ T cells but not CD4+ T cells reduced retinal neovascularization and vascular leakage. Using reporter mice expressing gfp (green fluorescence protein) in CD8+ T cells, these cells were localized near neovascular tufts in the retina, confirming that CD8+ T cells contribute to the disease. Furthermore, the adoptive transfer of CD8+ T cells deficient in TNF (tumor necrosis factor), IFNγ (interferon gamma), Prf (perforin), or GzmA/B (granzymes A/B) into immunocompetent Rag1-/- mice revealed that CD8+ T cells mediate retinal vascular disease via these factors, with TNF influencing all aspects of vascular pathology. The pathway by which CD8+ T cells migrate into the retina was identified as CXCR3 (C-X-C motif chemokine receptor 3) with the CXCR3 blockade reducing the number of CD8+ T cells within the retina and retinal vascular disease. CONCLUSIONS: We discovered that CXCR3 is central to the migration of CD8+ T cells into the retina as the CXCR3 blockade reduced the number of CD8+ T cells within the retina and vasculopathy. This research identified an unappreciated role for CD8+ T cells in retinal inflammation and vascular disease. Reducing CD8+ T cells via their inflammatory and recruitment pathways is a potential treatment for neovascular retinopathies.


Assuntos
Doenças Retinianas , Doenças Vasculares , Animais , Camundongos , Linfócitos T CD8-Positivos/metabolismo , Neovascularização Patológica , Retina/metabolismo , Doenças Retinianas/metabolismo , Interferon gama/metabolismo , Doenças Vasculares/patologia , Camundongos Endogâmicos C57BL
6.
BMC Ophthalmol ; 24(1): 385, 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39218852

RESUMO

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.


Assuntos
Angiofluoresceinografia , Neovascularização Retiniana , Retinite , Humanos , Feminino , Adulto , Neovascularização Retiniana/diagnóstico , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/etiologia , Retinite/diagnóstico , Retinite/microbiologia , Retinite/tratamento farmacológico , Angiofluoresceinografia/métodos , Tomografia de Coerência Óptica/métodos , Infecções Oculares Bacterianas/diagnóstico , Infecções Oculares Bacterianas/microbiologia , Infecções Oculares Bacterianas/tratamento farmacológico , Infecções por Rickettsia/diagnóstico , Infecções por Rickettsia/complicações , Infecções por Rickettsia/tratamento farmacológico , Infecções por Rickettsia/microbiologia , Rickettsia conorii , Fundo de Olho , Acuidade Visual , Inibidores da Angiogênese/uso terapêutico , Inibidores da Angiogênese/administração & dosagem , Injeções Intravítreas , Doxiciclina/uso terapêutico , Bevacizumab/uso terapêutico
7.
BMC Genomics ; 24(1): 327, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37322431

RESUMO

BACKGROUND: Retinal neovascularization (RNV) is a leading cause of blindness worldwide. Long non-coding RNA (lncRNA) and competing endogenous RNA (ceRNA) regulatory networks play vital roles in angiogenesis. The RNA-binding protein galectin-1 (Gal-1) participates in pathological RNV in oxygen-induced retinopathy mouse models. However, the molecular associations between Gal-1 and lncRNAs remain unclear. Herein, we aimed to explore the potential mechanism of action of Gal-1 as an RNA-binding protein. RESULTS: A comprehensive network of Gal-1, ceRNAs, and neovascularization-related genes was constructed based on transcriptome chip data and bioinformatics analysis of human retinal microvascular endothelial cells (HRMECs). We also conducted functional enrichment and pathway enrichment analyses. Fourteen lncRNAs, twenty-nine miRNAs, and eleven differentially expressed angiogenic genes were included in the Gal-1/ceRNA network. Additionally, the expression of six lncRNAs and eleven differentially expressed angiogenic genes were validated by qPCR in HRMECs with or without siLGALS1. Several hub genes, such as NRIR, ZFPM2-AS1, LINC0121, apelin, claudin-5, and C-X-C motif chemokine ligand 10, were found to potentially interact with Gal-1 via the ceRNA axis. Furthermore, Gal-1 may be involved in regulating biological processes related to chemotaxis, chemokine-mediated signaling, the immune response, and the inflammatory response. CONCLUSIONS: The Gal-1/ceRNA axis identified in this study may play a vital role in RNV. This study provides a foundation for the continued exploration of therapeutic targets and biomarkers associated with RNV.


Assuntos
MicroRNAs , RNA Longo não Codificante , Neovascularização Retiniana , Animais , Humanos , Camundongos , Quimiocinas , Células Endoteliais , Galectina 1/genética , Redes Reguladoras de Genes , MicroRNAs/genética , Neovascularização Retiniana/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética
8.
J Vasc Res ; 60(4): 183-192, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37660689

RESUMO

OBJECTIVE: The aim of the study was to evaluate the effect of the RhoA/ROCK inhibitor Fasudil on retinal neovascularization (NV) in vivo and angiogenesis in vitro. METHODS: C57BL/6 was used to establish an OIR model. First, RhoA/ROCK expression was first examined and compared between OIR and healthy controls. Then, we evaluated the effect of Fasudil on pathological retinal NV. Whole-mount retinal staining was performed. The percentage of NV area, the number of neovascular tufts (NVT), and branch points (BP) were quantified. Finally, human umbilical vein endothelial cells (HUVECs) were used to investigate the effect of Fasudil on angiogenesis. RESULTS: Real-time PCR and Western blotting showed that ROCK expression in retinal tissue was statistically upregulated in OIR. Furthermore, we found that Fasudil attenuated the percentage of NV area, the number of NVT, and BP significantly. In addition, Fasudil could suppress the proliferation and migration of HUVECs induced by VEGF. CONCLUSIONS: RhoA/ROCK might be involved in the pathogenesis of OIR. And its inhibitor Fasudil could suppress retinal NV in vivo and angiogenesis in vitro. Fasudil may be a potential treatment strategy for retinal vascular diseases.


Assuntos
Neovascularização Retiniana , Humanos , Animais , Camundongos , Neovascularização Retiniana/genética , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Neovascularização Patológica/patologia , Retina/metabolismo , Células Endoteliais da Veia Umbilical Humana , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
9.
Pharmacol Res ; 187: 106617, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36535572

RESUMO

Retinal neovascularization, or pathological angiogenesis in the retina, is a leading cause of blindness in developed countries. Transforming growth factor-ß-activated kinase 1 (TAK1) is a mitogen-activated protein kinase kinase kinase (MAPKKK) activated by TGF-ß1 and other proinflammatory cytokines. TAK1 is also a key mediator of proinflammatory signals and plays an important role in maintaining vascular integrity upon proinflammatory cytokine stimulation such as TNFα. However, its role in pathological angiogenesis, particularly in retinal neovascularization, remains unclear. Here, we investigate the regulatory role of TAK1 in human endothelial cells responding to inflammatory stimuli and in a rat model of oxygen-induced retinopathy (OIR) featured retinal neovascularization. Using TAK1 knockout human endothelial cells that subjected to inflammatory stimuli, transcriptome analysis revealed that TAK1 is required for activation of NFκB signaling and mediates its downstream gene expression related to endothelial activation and angiogenesis. Moreover, pharmacological inhibition of TAK1 by 5Z-7-oxozeaenol attenuated angiogenic activities of endothelial cells. Transcriptome analysis also revealed enrichment of TAK1-mediated NFκB signaling pathway in the retina of OIR rats and retinal neovascular membrane from patients with proliferative diabetic retinopathy. Intravitreal injection of 5Z-7-oxozeaenol significantly reduced hypoxia-induced inflammation and microglial activation, thus attenuating aberrant retinal angiogenesis in OIR rats. Our data suggest that inhibition of TAK1 may have therapeutic potential for the treatment of retinal neovascular pathologies.


Assuntos
Doenças Retinianas , Neovascularização Retiniana , Animais , Humanos , Camundongos , Ratos , Citocinas/uso terapêutico , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Lactonas/uso terapêutico , Camundongos Endogâmicos C57BL , Neovascularização Patológica/patologia , NF-kappa B , Oxigênio , Doenças Retinianas/patologia , Neovascularização Retiniana/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 42(4): 407-427, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35236105

RESUMO

BACKGROUND: The major aim of this study is to investigate whether CDC6 (cell division cycle 6), a replication origin recognition complex component, plays a role in retinal neovascularization, and if so, to explore the underlying mechanisms. METHODS: In this study, we used a variety of approaches including cellular and moleculer biological methodologies as well as global and tissue-specific knockout mice in combination with an oxygen-induced retinopathy model to study the role of CDC6 in retinal neovascularization. RESULTS: VEGFA (vascular endothelial growth factor A)-induced CDC6 expression in a time-dependent manner in human retinal microvascular endothelial cells. In addition, VEGFA-induced CDC6 expression was dependent on PLCß3 (phospholipase Cß3)-mediated NFATc1 (nuclear factor of activated T cells c1) activation. Furthermore, while siRNA-mediated depletion of PLCß3, NFATc1, or CDC6 levels blunted VEGFA-induced human retinal microvascular endothelial cell angiogenic events such as proliferation, migration, sprouting, and tube formation, CDC6 overexpression rescued these effects in NFATc1-deficient mouse retinal microvascular endothelial cells. In accordance with these observations, global knockdown of PLCß3 or endothelial cell-specific deletion of NFATc1 or siRNA-mediated depletion of CDC6 levels substantially inhibited oxygen-induced retinopathy-induced retinal sprouting and neovascularization. In addition, retroviral-mediated overexpression of CDC6 rescued oxygen-induced retinopathy-induced retinal neovascularization from inhibition in PLCß3 knockout mice and in endothelial cell-specific NFATc1-deficient mice. CONCLUSIONS: The above observations clearly reveal that PLCß3-mediated NFATc1 activation-dependent CDC6 expression plays a crucial role in VEGFA/oxygen-induced retinopathy-induced retinal neovascularization.


Assuntos
Doenças Retinianas , Neovascularização Retiniana , Animais , Ciclo Celular , Proteínas de Ciclo Celular , Células Endoteliais , Camundongos , Camundongos Knockout , Proteínas Nucleares , Oxigênio , RNA Interferente Pequeno , Neovascularização Retiniana/genética , Fatores de Transcrição , Fator A de Crescimento do Endotélio Vascular
11.
Int J Med Sci ; 20(2): 254-261, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36794165

RESUMO

Ischemia-induced pathological neovascularization in the retina is a leading cause of blindness in various age groups. The purpose of the current study was to identify the involvement of circular RNAs (circRNAs) methylated by N6-methyladenosine (m6A), and predict their potential roles in oxygen-induced retinopathy (OIR) in mice. Methylation assessment via microarray analysis indicated that 88 circRNAs were differentially modified by m6A methylation, including 56 hyper-methylated circRNAs and 32 hypo-methylated circRNAs. Gene ontology enrichment analysis predicted that the enriched host genes of the hyper-methylated circRNAs were involved in cellular process, cellular anatomical entity, and protein binding. Host genes of the hypo-methylated circRNAs were enriched in the regulation of cellular biosynthetic process, the nucleus, and binding. According to the Kyoto Encyclopedia of Genes and Genomes analysis, those host genes were involved in the pathways of selenocompound metabolism, salivary secretion, and lysine degradation. MeRIP-qPCR verified significant alterations in m6A methylation levels of mmu_circRNA_33363, mmu_circRNA_002816, and mmu_circRNA_009692. In conclusion, the study revealed the m6A modification alterations in OIR retinas, and the findings above shed light on the potential roles of m6A methylation in circRNA regulatory functions in the pathogenesis of ischemia-induced pathological retinal neovascularization.


Assuntos
RNA Circular , Neovascularização Retiniana , Animais , Camundongos , RNA Circular/genética , RNA Circular/metabolismo , RNA/genética , RNA/metabolismo , Neovascularização Retiniana/genética , Perfilação da Expressão Gênica , Isquemia/complicações , Isquemia/genética
12.
Graefes Arch Clin Exp Ophthalmol ; 261(3): 615-626, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36171459

RESUMO

PURPOSE: Retinal neovascularization (RNV) is an intractable pathological hallmark of numerous ocular blinding diseases, including diabetic retinopathy, retinal vein occlusion, and retinopathy of prematurity. However, current therapeutic methods have potential side effects and limited efficacy. Thus, further studies on the pathogenesis of RNV-related disorders and novel therapeutic targets are critically required. Long non-coding RNAs (lncRNAs) have various functions and participate in almost all biological processes in living cells, such as translation, transcription, signal transduction, and cell cycle control. In addition, recent research has demonstrated critical modulatory roles of various lncRNAs in RNV. In this review, we summarize current knowledge about the expression and regulatory functions of lncRNAs related to the progression of pathological RNV. METHODS: We searched databases such as PubMed and Web of Science to gather and review information from the published literature. CONCLUSIONS: In general, lncRNA MEG3 attenuates RNV, thus protecting the retina from excessive and dysregulated angiogenesis under high glucose stress. In contrast, lncRNAs MALAT1, MIAT, ANRIL, HOTAIR, HOTTIP, and SNHG16, have been identified as causative molecules in the pathological progression of RNV. Comprehensive and in-depth studies of the roles of lncRNAs in RNV indicate that targeting lncRNAs may be an alternative therapeutic approach in the near future, enabling new options for attenuating RNV progression and treating RNV-related retinal diseases.


Assuntos
RNA Longo não Codificante , Neovascularização Retiniana , Recém-Nascido , Humanos , Neovascularização Retiniana/etiologia , RNA Longo não Codificante/genética , Oxigênio/metabolismo , Retina/patologia , Neovascularização Patológica/genética
13.
Graefes Arch Clin Exp Ophthalmol ; 261(2): 339-344, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36303061

RESUMO

PURPOSE: Xephilio OCT-S1 can capture single-acquisition 23 × 20-mm wide-field swept-source optical coherence tomography angiography (SS-OCTA) images and high-resolution images using artificial intelligence. We aimed to evaluate the ability of wide-field SS-OCTA in the detection of retinal neovascularizations (NVs) in eyes with proliferative diabetic retinopathy (PDR). METHODS: This retrospective study included 64 eyes of 36 patients (age, 57 ± 10 years; 10 female, 26 male) with PDR. All patients underwent a comprehensive ophthalmological examination, including fluorescein angiography (FA), as well as fovea- and disc-centered 23 × 20-mm OCTA imaging (A-scan/B-scan, 928/807). We compared and examined the number of NV sites identified using conventional methods (merging the findings from biomicroscopy/color fundus photography, FA) and the number of NV sites identified using vitreoretinal interface and superficial retinal slabs of wide-field SS-OCTA images, including the position of NVs (nasal upper, nasal lower, temporal upper, temporal lower, or disc). RESULTS: We identified 168 NVs (32/40/45/35/16, in the abovementioned order) using the conventional method. Fovea-centered 23 × 20-mm OCTA images revealed 162 (96%) NVs (27/39/45/35/16). This method tended to miss nasal NV. In contrast, disc-centered 23 × 20-mm OCTA images identified nearly all NVs, detecting 166 (99%) NVs (32/40/44/34/16) in total. All NVs could be visualized using two wide-field OCTA images: fovea- and disc-centered. CONCLUSION: Wide-field (23 × 20 mm) SS-OCTA-especially disc-centered-using Xephilio OCT-S1 identified nearly all NVs in eyes with PDR, with a single acquisition, thereby demonstrating its potential clinical application.


Assuntos
Diabetes Mellitus , Retinopatia Diabética , Neovascularização Retiniana , Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Neovascularização Retiniana/diagnóstico , Retinopatia Diabética/complicações , Retinopatia Diabética/diagnóstico , Vasos Retinianos , Tomografia de Coerência Óptica/métodos , Estudos Retrospectivos , Inteligência Artificial , Angiofluoresceinografia/métodos
14.
Int J Mol Sci ; 24(14)2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37511525

RESUMO

MicroRNA (miRNA) is a non-coding RNA that can regulate the expression of many target genes, and it is widely involved in various important physiological activities. MiR-124-3p was found to associate with the normal development of retinal vessels in our previous study, but the mechanism of its anti-angiogenic effect on pathological retinal neovascularization still needed to be explored. Therefore, this study aimed to investigate the effect and mechanism of miR-124-3p on retinal neovascularization in mice with oxygen-induced retinopathy (OIR). Here, we found that intravitreal injection of miR-124-3p agomir attenuated pathological retinal neovascularization in OIR mice. Moreover, miR-124-3p preserved the astrocytic template, inhibited reactive gliosis, and reduced the inflammatory response as well as necroptosis. Furthermore, miR-124-3p inhibited the signal transducer and activator of transcription 3 (STAT3) pathway and decreased the expression of hypoxia-inducible factor-1α and vascular endothelial growth factor. Taken together, our results revealed that miR-124-3p inhibited retinal neovascularization and neuroglial dysfunction by targeting STAT3 in OIR mice.


Assuntos
MicroRNAs , Neovascularização Retiniana , Animais , Camundongos , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Neuroglia/metabolismo , Oxigênio/efeitos adversos , Oxigênio/metabolismo , Neovascularização Retiniana/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Biochem Biophys Res Commun ; 617(Pt 2): 69-76, 2022 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-35691117

RESUMO

Diabetic retinopathy (DR) is a key reason for legal blindness worldwide. Currently, it is urgently necessary to determine the etiology and pathological molecular mechanism of DR to search for resultful therapies. Dickkopf-1 (DKK1) is inhibitive for canonical Wnt signaling via negative feedback, and has been reported as a biomarker for DR. However, the related mechanisms are still unclear. In this work, our data showed that DKK1 was decreased in the vitreous tissues at an early stage of diabetes triggered by streptozotocin (STZ) injection in rats. We subsequently found that DKK1 intravitreal injection significantly ameliorated the physiological function of retina in STZ-challenged rats, accompanied by improved retinal structure. Surprisingly, our results indicated that DKK1 injection remarkably suppressed PANoptosis in retinal tissues of STZ-challenged rats with DR, as proved by ameliorated pyroptosis, apoptosis and necroptosis, which were mainly through the blockage of cleaved Gasdermin-D (GSDMD), Caspase-3 and receptor-interacting protein kinase-3 (RIPK3). Additionally, Wnt signaling including the expression of Wnt, ß-catenin and LDL receptor-related protein 5/6 (LRP5/6) was also highly prohibited in retina of DKK1-injected rats with DR. Furthermore, retinal neovascularization and acellular vessel in DR rats were also considerably abolished after DKK1 injection, accompanied by reduced expression levels of retinal vascular endothelial growth factor (VEGF), matrix metalloproteinase-2 (MMP2) and matrix metalloproteinase-9 (MMP9). More in vitro experiments showed that DKK1 treatment markedly repressed the proliferative and migratory ability of endothelial cells via inhibiting angiogenesis-related molecules. Together, all our results broaden the knowledge of the correlation between DKK1 and DR, and then provide a novel therapeutic strategy for the suppression of management of DR.


Assuntos
Diabetes Mellitus Experimental , Retinopatia Diabética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neovascularização Retiniana , Animais , Diabetes Mellitus Experimental/metabolismo , Retinopatia Diabética/metabolismo , Células Endoteliais/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Ratos , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/prevenção & controle , Estreptozocina , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Microvasc Res ; 141: 104315, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35007537

RESUMO

BACKGROUND: Retinal neovascularization (RN), a major cause of blindness occurring in multiple types of ophthalmic diseases, is closely associated with hypoxic conditions. However, the underlying pathological mechanisms of RN have not been fully elucidated. BTG2 is anti-proliferative factor. The up-stream of BTG2 gene within 3000 bp expresses a long non-coding RNA, LNC01136. METHODS: we initially compared the expression of BTG2 and LNC01136 in human retinal microvascular endothelial cells (hRMECs) with other eye-associated cells, including Muller cells, ARPE19 cells and RGC-5, in response to a hypoxia mimetic agent (CoCl2). FISH and PCR tests were performed to determine the enrichment of LNC01136 in different cellular components. LNC01136 were overexpressed or knockdown to determine the effect on BTG2 expression. Finally, ChIP, RIP and Co-IP assays were performed to determine the interaction among BTG2, HIF-1α, LNC01136 and CNOT7. RESULTS: After the treatment with CoCl2, expression levels of BTG2 and LNC01136 were strongly induced in Muller cells, ARPE19 cells and RGC-5, but weakly in hRMECs. LNC01136 is prominently located in cell nucleus and aids HIF-1α to enhance transcription of BTG2, which consequently inhibits cell growth. The anti-proliferative effect of BTG2 is probably associated to the interaction with CNOT7 and the regulation of multiple cell cycle-related proteins. CONCLUSIONS: This study revealed that LNC01136 is a cell growth suppressor by recruiting HIF-1α to induce BTG2 expression. However the low expression of LNC01136 in hRMECs compared to other eye-associated cells promoted hRMECs' proliferation, which is probably a cause of RN under hypoxia.


Assuntos
Proteínas Imediatamente Precoces , Neovascularização Retiniana , Hipóxia Celular/fisiologia , Proliferação de Células , Células Endoteliais/metabolismo , Humanos , Hipóxia/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Retina/metabolismo , Neovascularização Retiniana/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
17.
J Nanobiotechnology ; 20(1): 174, 2022 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-35366885

RESUMO

BACKGROUND: Pathological retinal angiogenesis resulting from a variety of ocular diseases including oxygen induced retinopathy, diabetic retinopathy and ocular vein occlusion, is one of the major reasons for vision loss, yet the therapeutic option is limited. Multiple nanoparticles have been reported to alleviate angiogenic retinopathy. However, the adverse effect cannot be ignored due to the relatively large scale. Graphene quantum dots (GQDs) have shown potential in drug delivery and have been proved biocompatible. In this study, Graphene quantum dots are extensively investigated for their application in angiogenic retinopathy therapy. RESULTS: We showed that GQDs were biocompatible nanomaterials in vitro and in vivo. The nanoparticles have a dose-dependent inhibitory effect on proliferation, migration, tube formation and sprouting of human umbilical vein endothelial cells (HUVECs). Further data show that GQDs could inhibit pathological retinal neovascularization in an oxygen-induced retinopathy (OIR) model. The data of RNA sequencing suggested that periostin is involved in this process. GQDs inhibit the expression of periostin via STAT3, and further regulated cell cycle-related protein levels through ERK pathway. The signaling pathway was conformed in vivo using OIR mouse model. CONCLUSIONS: The present study indicated that GQDs could be a biocompatible anti-angiogenic nanomedicine in the treatment of pathological retinal neovascularization via disrupting periostin/ERK pathway and subsequent cell cycle.


Assuntos
Grafite , Pontos Quânticos , Doenças Retinianas , Animais , Proliferação de Células , Células Cultivadas , Grafite/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Pontos Quânticos/uso terapêutico , Transdução de Sinais
18.
Graefes Arch Clin Exp Ophthalmol ; 260(2): 553-559, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34499246

RESUMO

PURPOSE: This study aims to report the long-term outcomes of uveitis-associated optic disc and epiretinal neovascularization (NV) treated with immunomodulatory therapy alone. METHODS: This is a retrospective, multi-center chart review conducted at Northwestern University (Chicago, IL) and San Raffaele Scientific Institute (Milan, Italy) from 2014 to 2021 of patients with optic disc and/or retinal neovascularization associated with uveitis. The data collected included age at the time of NV detection, gender, medications, and follow-up period. Imaging was reviewed if available. RESULTS: Eight eyes of six patients were identified. The mean age was 22 years (range 10-52 years); the median follow-up was 3 years (range 6 months to 7 years). All eyes presented with active NV at the time of uveitis onset; 7 eyes were treatment-naïve. None had clinical or angiographic evidence of retinal ischemia. All patients received a variable combination of local steroids, systemic steroids, and systemic immunosuppression. Complete resolution of uveitic NV occurred in all eyes within a median of 8 weeks (ranging 2-20 weeks) from initiating treatment. No NV recurrence was noted. CONCLUSION: Immunomodulatory therapy alone may be successful in achieving long-term control of uveitis-associated NV, without the use of destructive measures.


Assuntos
Disco Óptico , Neovascularização Retiniana , Uveíte , Adolescente , Adulto , Criança , Seguimentos , Humanos , Imunomodulação , Pessoa de Meia-Idade , Estudos Retrospectivos , Uveíte/diagnóstico , Uveíte/tratamento farmacológico , Adulto Jovem
19.
Lasers Surg Med ; 54(5): 747-757, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35320609

RESUMO

OBJECTIVES: Retinal neovascularization (RNV) is the growth of abnormal microvessels on the retinal surface and into the vitreous, which can lead to severe vision loss. By combining relatively low-intensity ultrasound and nanosecond-pulse-duration laser, we developed a novel treatment method, namely photo-mediated ultrasound therapy (PUT), which holds a potential to remove RNV with minimal or no damage to the adjacent tissues. METHODS: RNV was created in both albino and pigmented rabbits (n = 10) through a single intravitreal injection with DL-α-aminoadipic acid. RNV was treated with PUT 8 weeks postinjection. After PUT treatment, animals were evaluated longitudinally for up to 6 weeks. Treatment outcomes were evaluated through fundus photography, red-free fundus photography, fluorescein angiography (FA), and histopathology. RESULTS: In both albino and pigmented rabbits, there were no leakage in the treatment area immediately after PUT treatment as demonstrated by FA, indicating the cessation of blood perfusion of the RNV in the treated area. The fluorescence leakage did not recover in albino rabbits during the 6-week posttreatment monitoring period, and only 9.9 ± 9.8% of the neovascularization remained at the end of 6 weeks. In the pigmented rabbits, the fluorescence leakage partially returned, but the level of leakage decreased over time during the 6-week posttreatment monitoring period, and only 10.8 ± 9.8% of the neovascularization remained at the end of 6 weeks. Histology demonstrated removal of vasculature without damage to the surrounding neurosensory retina. CONCLUSIONS: These results demonstrate that PUT could precisely remove RNV without damage to the surrounding neurosensory retina in both rabbit strains.


Assuntos
Neovascularização Retiniana , Terapia por Ultrassom , Animais , Angiofluoresceinografia , Injeções Intravítreas , Coelhos , Retina/diagnóstico por imagem , Retina/patologia , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/patologia
20.
Int J Mol Sci ; 23(15)2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35955664

RESUMO

Retinopathy of prematurity (ROP) is a rare proliferative ocular disorder in preterm infants. Because of the advancements in neonatal care, the incidence of ROP has increased gradually. Now, ROP is one of the leading causes of blindness in children. Preterm infants with immature retinal development are exposed to supplemental oxygen inside an incubator until their cardiopulmonary system is adequately developed. Once they are returned to room air, the relatively low oxygen level stimulates various angiogenesis factors initiating retinal neovascularization. If patients with ROP are not offered adequate and timely treatment, they can experience vision loss that may ultimately lead to permanent blindness. Although laser therapy and anti-vascular endothelial growth factor agents are widely used to treat ROP, they have limitations. Thus, it is important to identify novel therapeutics with minimal adverse effects for the treatment of ROP. To date, various pharmacologic and non-pharmacologic therapies have been assessed as treatments for ROP. In this review, the major molecular factors involved in the pathogenesis of ROP, currently offered therapies, therapies under investigation, and emerging novel therapeutics of ROP are discussed.


Assuntos
Doenças do Prematuro , Retinopatia da Prematuridade , Cegueira/complicações , Cegueira/tratamento farmacológico , Criança , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Oxigênio , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA