Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 706
Filtrar
1.
Sci Rep ; 14(1): 15647, 2024 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-38977744

RESUMO

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.


Assuntos
Inibidores da Angiogênese , Ranibizumab , Retinopatia da Prematuridade , Humanos , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Ranibizumab/administração & dosagem , Ranibizumab/uso terapêutico , Masculino , Feminino , Recém-Nascido , Estudos Retrospectivos , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/uso terapêutico , Resultado do Tratamento , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/patologia , Injeções Intravítreas , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Lactente , Recém-Nascido Prematuro
2.
Angiogenesis ; 27(3): 423-440, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38709389

RESUMO

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.


Assuntos
Oxigênio , Retinopatia da Prematuridade , Animais , Retinopatia da Prematuridade/patologia , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/metabolismo , Camundongos , Anticorpos Monoclonais Humanizados/farmacologia , Camundongos Endogâmicos C57BL , Animais Recém-Nascidos , Modelos Animais de Doenças , Humanos , Hiperóxia/patologia , Hiperóxia/complicações , Retina/patologia , Retina/metabolismo , Retina/efeitos dos fármacos , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Camundongos Transgênicos , Neovascularização Retiniana/patologia , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/tratamento farmacológico , Microglia/patologia , Microglia/metabolismo , Microglia/efeitos dos fármacos
3.
Immunology ; 173(1): 141-151, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38804253

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Flavonoides , Hexoquinase , Microglia , Oxigênio , Neovascularização Retiniana , Retinopatia da Prematuridade , Transdução de Sinais , Serina-Treonina Quinases TOR , Animais , Hexoquinase/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Oxigênio/metabolismo , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/patologia , Camundongos , Humanos , Neovascularização Retiniana/tratamento farmacológico , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos Endogâmicos C57BL , Movimento Celular/efeitos dos fármacos
4.
Angiogenesis ; 27(3): 379-395, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38483712

RESUMO

Pathological retinal angiogenesis profoundly impacts visual function in vascular eye diseases, such as retinopathy of prematurity (ROP) in preterm infants and age-related macular degeneration in the elderly. While the involvement of photoreceptors in these diseases is recognized, the underlying mechanisms remain unclear. This study delved into the pivotal role of photoreceptors in regulating abnormal retinal blood vessel growth using an oxygen-induced retinopathy (OIR) mouse model through the c-Fos/A disintegrin and metalloprotease 17 (Adam17) axis. Our findings revealed a significant induction of c-Fos expression in rod photoreceptors, and c-Fos depletion in these cells inhibited pathological neovascularization and reduced blood vessel leakage in the OIR mouse model. Mechanistically, c-Fos directly regulated the transcription of Adam17 a shedding protease responsible for the production of bioactive molecules involved in inflammation, angiogenesis, and cell adhesion and migration. Furthermore, we demonstrated the therapeutic potential by using an adeno-associated virus carrying a rod photoreceptor-specific short hairpin RNA against c-fos which effectively mitigated abnormal retinal blood vessel overgrowth, restored retinal thickness, and improved electroretinographic (ERG) responses. In conclusion, this study highlights the significance of photoreceptor c-Fos in ROP pathology, offering a novel perspective for the treatment of this disease.


Assuntos
Proteína ADAM17 , Proteínas Proto-Oncogênicas c-fos , Neovascularização Retiniana , Animais , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Neovascularização Retiniana/genética , Proteína ADAM17/metabolismo , Proteína ADAM17/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Camundongos , Humanos , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/patologia , Retinopatia da Prematuridade/genética , Camundongos Endogâmicos C57BL , Transcrição Gênica , Regulação da Expressão Gênica , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Modelos Animais de Doenças , Angiogênese
5.
Int J Mol Med ; 53(5)2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38551157

RESUMO

Macrophages form a crucial component of the innate immune system, and their activation is indispensable for various aspects of immune and inflammatory processes, tissue repair, and maintenance of the balance of the body's state. Macrophages are found in all ocular tissues, spanning from the front surface, including the cornea, to the posterior pole, represented by the choroid/sclera. The neural retina is also populated by specialised resident macrophages called microglia. The plasticity of microglia/macrophages allows them to adopt different activation states in response to changes in the tissue microenvironment. When exposed to various factors, microglia/macrophages polarise into distinct phenotypes, each exhibiting unique characteristics and roles. Furthermore, extensive research has indicated a close association between microglia/macrophage polarisation and the development and reversal of various intraocular diseases. The present article provides a review of the recent findings on the association between microglia/macrophage polarisation and ocular pathological processes (including autoimmune uveitis, optic neuritis, sympathetic ophthalmia, retinitis pigmentosa, glaucoma, proliferative vitreoretinopathy, subretinal fibrosis, uveal melanoma, ischaemic optic neuropathy, retinopathy of prematurity and choroidal neovascularization). The paradoxical role of microglia/macrophage polarisation in retinopathy of prematurity is also discussed. Several studies have shown that microglia/macrophages are involved in the pathology of ocular diseases. However, it is required to further explore the relevant mechanisms and regulatory processes. The relationship between the functional diversity displayed by microglia/macrophage polarisation and intraocular diseases may provide a new direction for the treatment of intraocular diseases.


Assuntos
Microglia , Retinopatia da Prematuridade , Recém-Nascido , Humanos , Microglia/patologia , Retinopatia da Prematuridade/patologia , Retina/patologia , Macrófagos , Fenótipo
6.
Am J Ophthalmol ; 260: 190-199, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38141904

RESUMO

PURPOSE: Experimental studies provide evidence that regulation of VEGF receptor-2 signaling in endothelial cells orders cell divisions and extends developmental angiogenesis while inhibiting pathologic intravitreal angiogenesis and has relevance to retinopathy of prematurity (ROP). We tested the hypothesis that intravitreal anti-VEGF would extend vascularization into peripheral avascular retina in human type 1 ROP compared with controls. DESIGN: Retrospective, nonrandomized treatment comparison. METHODS: The study was conducted at an academic institution, with the study population comprising all premature infants screened for ROP from January 2019 through December 2022. The experimental group included type 1 ROP treated with bilateral bevacizumab (0.25 mg) and had adequate fundus imaging by a certified ophthalmic photographer at 2 examinations: within 2 weeks of treatment and 1-3 weeks later. A control group included gestational age- and birthweight-matched infants with ROP less severe than type 1 ROP. The main outcome measure was extent of temporal retinal vasculature measured by a masked analyst between treated and control eyes. Paired and nonpaired t tests were used. RESULTS: Of 382 screened infants, 34 developed type 1 ROP; 11 comprised the experimental group and 11 the control group. At baseline, there was a trend toward shorter temporal vascular extent in treatment compared with control groups (3667±547 vs 4262±937 pixels, 95% CI -1277, 88; P = .084) but no difference between groups at follow-up (P = .945). Vascular extension was significantly greater in the treatment than control (872±521 vs 253±151 pixels, 95% CI 262, 977; P = .003), showing catch-up growth. CONCLUSIONS: This clinical evidence supports laboratory-based studies that regulation of VEGF using an intravitreal anti-VEGF agent increases developmental angiogenesis into the peripheral avascular retina while inhibiting pathologic intravitreal angiogenesis in ROP.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Recém-Nascido , Lactente , Humanos , Inibidores da Angiogênese/uso terapêutico , Fator A de Crescimento do Endotélio Vascular , Retinopatia da Prematuridade/diagnóstico , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Células Endoteliais/patologia , Estudos Retrospectivos , Injeções Intravítreas , Bevacizumab/uso terapêutico , Recém-Nascido Prematuro , Idade Gestacional , Neovascularização Retiniana/diagnóstico , Neovascularização Retiniana/tratamento farmacológico , Retina/patologia
7.
Cells ; 12(20)2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37887312

RESUMO

The progression to fibrosis and traction in retinopathy of prematurity (ROP) and other ischemic retinopathies remains an important clinical and surgical challenge, necessitating a comprehensive understanding of its pathogenesis. Fibrosis is an unbalanced deposition of extracellular matrix components responsible for scar tissue formation with consequent tissue and organ impairment. Together with retinal traction, it is among the main causes of retinal detachment and vision loss. We capitalize on the Limited Hyperoxia Induced Retinopathy (LHIPR) model, as it reflects the more advanced pathological phenotypes seen in ROP and other ischemic retinopathies. To model LHIPR, we exposed wild-type C57Bl/6J mouse pups to 65% oxygen from P0 to P7. Then, the pups were returned to room air to recover until later endpoints. We performed histological and molecular analysis to evaluate fibrosis progression, angiogenesis, and inflammation at several time points, from 1.5 months to 9 months. In addition, we performed in vivo retinal imaging by optical coherence tomography (OCT) or OCT Angiography (OCTA) to follow the fibrovascular progression in vivo. Although the retinal morphology was relatively preserved, we found a progressive increase in preretinal fibrogenesis over time, up to 9 months of age. We also detected blood vessels in the preretinal space as well as an active inflammatory process, altogether mimicking advanced preretinal fibrovascular disease in humans.


Assuntos
Hiperóxia , Neovascularização Retiniana , Retinopatia da Prematuridade , Vitreorretinopatia Proliferativa , Animais , Camundongos , Fibrose , Hiperóxia/complicações , Inflamação/patologia , Isquemia/patologia , Camundongos Endogâmicos C57BL , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/patologia , Vasos Retinianos , Retinopatia da Prematuridade/induzido quimicamente , Retinopatia da Prematuridade/complicações , Retinopatia da Prematuridade/patologia , Vitreorretinopatia Proliferativa/patologia
8.
Am J Pathol ; 193(12): 2001-2016, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37673326

RESUMO

Bronchopulmonary dysplasia (BPD), also called chronic lung disease of immaturity, afflicts approximately one third of all extremely premature infants, causing lifelong lung damage. There is no effective treatment other than supportive care. Retinopathy of prematurity (ROP), which impairs vision irreversibly, is common in BPD, suggesting a related pathogenesis. However, specific mechanisms of BPD and ROP are not known. Herein, a neonatal mouse hyperoxic model of coincident BPD and retinopathy was used to screen for candidate mediators, which revealed that granulocyte colony-stimulating factor (G-CSF), also known as colony-stimulating factor 3, was up-regulated significantly in mouse lung lavage fluid and plasma at postnatal day 14 in response to hyperoxia. Preterm infants with more severe BPD had increased plasma G-CSF. G-CSF-deficient neonatal pups showed significantly reduced alveolar simplification, normalized alveolar and airway resistance, and normalized weight gain compared with wild-type pups after hyperoxic lung injury. This was associated with a marked reduction in the intensity, and activation state, of neutrophilic and monocytic inflammation and its attendant oxidative stress response, and protection of lung endothelial cells. G-CSF deficiency also provided partial protection against ROP. The findings in this study implicate G-CSF as a pathogenic mediator of BPD and ROP, and suggest the therapeutic utility of targeting G-CSF biology to treat these conditions.


Assuntos
Displasia Broncopulmonar , Hiperóxia , Retinopatia da Prematuridade , Lactente , Recém-Nascido , Animais , Humanos , Camundongos , Displasia Broncopulmonar/patologia , Recém-Nascido Prematuro , Células Endoteliais/patologia , Pulmão/patologia , Hiperóxia/complicações , Retinopatia da Prematuridade/patologia , Fator Estimulador de Colônias de Granulócitos , Animais Recém-Nascidos
9.
Invest Ophthalmol Vis Sci ; 64(11): 8, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37540175

RESUMO

Purpose: SYVN1, a gene involved in endoplasmic reticulum-associated degradation, has been found to exert a protective effect by inhibiting inflammation in retinopathy. This study aimed to clarify whether SYVN1 is involved in the pathogenesis of retinopathy of prematurity (ROP) and its potential as a candidate for target therapy. Methods: Human retinal microvascular endothelial cells (hRMECs) and a mouse model of oxygen-induced retinopathy (OIR) were used to reveal the retinopathy development-associated protein expression and molecular mechanism. An adenovirus overexpressing SYVN1 or vehicle control was injected intravitreally at postnatal day 12 (P12), and the neovascular lesions were evaluated in retinal flatmounts with immunofluorescence staining, and hematoxylin and eosin staining at P17. Visual function was assessed by using electroretinogram (ERG). Results: Endogenous SYVN1 expression dramatically decreased in hRMECs under hypoxia and in ROP mouse retinas. SYVN1 regulated the signal transducer and activator of transcription 3 (STAT3)/vascular endothelial growth factor (VEGF) axis. SYVN1 overexpression promoted ubiquitination and degradation of STAT3, decreased the levels of phospho-STAT3, secretion of VEGF, and formation of neovascularization in hRMECs, which could be rescued by STAT3 activator treatment. In addition, SYVN1 overexpression prevented neovascularization and extended physiologic retinal vascular development in the retinal tissues of OIR mice without affecting retinal function. Conclusions: SYVN1 has a protective effect against OIR, and the molecular mechanisms are partly through SYVN1-mediated ubiquitination of STAT3 and the subsequent downregulation of VEGF. These findings strongly support our assumption that SYVN1 confers ROP resistance and may be a potentially novel pharmaceutical target against proliferative retinopathy.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Recém-Nascido , Animais , Camundongos , Humanos , Retinopatia da Prematuridade/patologia , Neovascularização Retiniana/metabolismo , Inibidores da Angiogênese/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator de Transcrição STAT3/metabolismo , Células Endoteliais/metabolismo , Degradação Associada com o Retículo Endoplasmático , Oxigênio/metabolismo , Neovascularização Patológica/metabolismo , Ubiquitinação , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Animais Recém-Nascidos , Ubiquitina-Proteína Ligases/genética
10.
Methods Mol Biol ; 2678: 27-36, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37326703

RESUMO

Diabetic retinopathy (DR) is one of the leading causes of vision loss worldwide. There are numerous animal models available for developing new ocular therapeutics and drug screening and to investigate the pathological processes involved in DR. Among those animal models, the oxygen-induced retinopathy (OIR) model, though originally developed as a model for retinopathy of prematurity, has also been used to investigate angiogenesis in proliferative DR with the phenomenon of ischemic avascular zones and pre-retinal neovascularization it demonstrated. Briefly, neonatal rodents are exposed to hyperoxia to induce vaso-obliteration. Upon removal from hyperoxia, hypoxia develops in the retina that eventually results in neovascularization. The OIR model is mostly used in small rodents such as mice and rats. Here, we describe a detailed experimental protocol of rat OIR model and the subsequent assessment of abnormal vasculature. By illustrating the vasculoprotective and anti-angiogenic activities of the treatment, OIR model might advance to a new platform for investigating novel ocular therapeutic strategies for DR.


Assuntos
Hiperóxia , Neovascularização Retiniana , Retinopatia da Prematuridade , Humanos , Recém-Nascido , Animais , Ratos , Camundongos , Oxigênio , Hiperóxia/complicações , Hiperóxia/patologia , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/patologia , Vasos Retinianos/patologia , Modelos Animais de Doenças , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/patologia , Retina/patologia , Camundongos Endogâmicos C57BL , Animais Recém-Nascidos
11.
Semin Ophthalmol ; 38(2): 124-133, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36536520

RESUMO

BACKGROUND: Retinal neovascularization is the major cause of vision loss that affects both adults and young children including premature babies. It has been a major pathology in several retinal diseases like age-related macular degeneration (AMD), diabetic retinopathy (DR) and retinopathy of prematurity (ROP). Current treatment modalities such as anti-VEGF therapy, laser are not suitable for every patient and response to these therapies is highly variable. Thus, there is a need to investigate newer therapeutic targets for DR, ROP and AMD, based on a clear understanding of disease pathology and regulatory mechanisms involved. METHOD: Appropriate articles published till February 2021 were extracted from PUBMED using keywords like ocular angiogenesis, DR, ROP, AMD, miRNA, mRNA, and cirMiRNA and containvaluable information regarding the involvement of miRNA in causing neovascularization. After compiling the list of miRNA regulating mRNA expression in angiogenesis and neovascularaization, their interactions were studied using online available tool MIENTURNET (http://userver.bio.uniroma1.it/apps/mienturnet/). The pathways involved in these processes were also predicted using the same tool. RESULTS: Most of the studies have explored potential targets like HIF1-α, PDGF, TGFß, FGF, etc., for their involvement in pathological angiogenesis in different retinal diseases. The regulatory role of microRNA (miRNA) has also been explored in various retinal ocular pathologies. This review highlights regulatory mechanism of cellular and circulatory miRNAs and their interactions with the genes involved in retinal neovascularization. The role of long noncoding RNA (ncRNA) in the regulation of genes involved in different pathways is also noteworthy and discussed in this review. CONCLUSION: This review highlights the potential regulatory mechanism/pathways involved in retinal neovascularization and its implications in retinal diseases and for identifying new drug targets.


Assuntos
Retinopatia Diabética , Degeneração Macular , MicroRNAs , Neovascularização Retiniana , Retinopatia da Prematuridade , Recém-Nascido , Criança , Humanos , Pré-Escolar , Neovascularização Retiniana/genética , Retina/patologia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Degeneração Macular/tratamento farmacológico , Retinopatia Diabética/genética , MicroRNAs/genética , MicroRNAs/uso terapêutico , RNA Mensageiro/uso terapêutico
12.
Exp Eye Res ; 226: 109347, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36502924

RESUMO

Retinopathy of prematurity (ROP) is a vision-threatening ocular disease that occurs in premature infants, but the underlying mechanism is still unclear. Since oxidative stress has been well documented in the ROP development, we aimed to investigate whether ferroptosis, a new type of cell death characterized by lipid peroxidation and iron overload, is also involved in ROP. We detected the lipid peroxidation, oxidative stress and the expression of ferroptosis markers in the retina of mouse model of oxygen-induced retinopathy. After ferroptosis inhibitor, ferrostatin-1, was administered by intravitreal injection, ferroptosis marker, lipid peroxidation, retinal vasculature and glial cell activation were examined. We found decreased expression of SLC7A11 and GPX4, increased expression of FTH1 and TFRC, as well as increase of lipid peroxidation in the retina of OIR mice. Ferrostatin-1 administration significantly reduced lipid peroxidation, and also reversed the change of ferroptosis marker. Neovascular area and avascular area were suppressed and the pathological vasculature changes including acellular vessels and ghost pericytes were decreased. Microglial cell and Müller cell activation was not evidently influenced by ferrostatin-1 treatment. Our findings suggest that ferroptosis is involved in the pathological angiogenesis and might be a promising target for ROP therapy.


Assuntos
Ferroptose , Neovascularização Patológica , Retinopatia da Prematuridade , Animais , Humanos , Recém-Nascido , Camundongos , Ferroptose/efeitos dos fármacos , Ferroptose/fisiologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Oxigênio/toxicidade , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Estresse Oxidativo
13.
Pediatr Res ; 93(5): 1250-1257, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-35986147

RESUMO

BACKGROUND: Retinopathy of prematurity (ROP) is the leading cause of blindness in infants, and elevation of HIF-1α through the PI3K/Akt and ERK pathways is implicated in ROP pathogenesis. The mechanism of action of propranolol in ROP remains controversial. We investigated the effect of propranolol on ROP and explored its potential mechanisms of action in an oxygen-induced retinopathy (OIR) mouse model. METHODS: OIR mice were first treated with propranolol intraperitoneally, and the retina integrity was measured by FITC-dextran and hematoxylin-eosin staining. The expression of HIF-1α, VEGF, and key signaling pathway proteins was determined using real-time PCR and western blotting. RESULTS: FITC-dextran staining showed that propranolol treatment reduced damage to retinal morphology in OIR mice. Mice treated with propranolol showed a reduced number of nuclei of vascular endothelial cells penetrating the inner limiting membrane of the retina, confirming the therapeutic effect of propranolol on ROP. Further analysis showed that HIF-1α and PI3K/Akt/ERK pathway protein levels were significantly elevated in OIR mice. In contrast, propranolol treatment downregulated the expression of these proteins, indicating that the PI3K/Akt/ERK/HIF-1α axis is associated with propranolol-induced ROP alleviation. CONCLUSIONS: Propranolol has a therapeutic function against ROP, likely through the downregulation of HIF-1α via the PI3K/Akt/ERK pathway. IMPACT: Propranolol can reduce the formation of abnormal retinal neovascularization in oxygen-induced retinopathy (OIR) models, and reduce leaking, tortuous, and abnormally expanding retinal blood vessels. Propranolol possibly improves OIR by inhibiting the activated ERK and HIF-1α pathways. Furthermore, propranolol may downregulate HIF-1α via the PI3K/Akt/ERK pathway to ameliorate retinopathy of prematurity. This study elucidated that the therapeutic effect of propranolol in OIR mice does not involve the VEGFR-2 pathway.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Humanos , Recém-Nascido , Animais , Camundongos , Propranolol/uso terapêutico , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células Endoteliais/metabolismo , Neovascularização Retiniana/metabolismo , Oxigênio/uso terapêutico , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças
14.
Elife ; 112022 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-36420952

RESUMO

At preterm birth, the retina is incompletely vascularized. Retinopathy of prematurity (ROP) is initiated by the postnatal suppression of physiological retinal vascular development that would normally occur in utero. As the neural retina slowly matures, increasing metabolic demand including in the peripheral avascular retina, leads to signals for compensatory but pathological neovascularization. Currently, only late neovascular ROP is treated. ROP could be prevented by promoting normal vascular growth. Early perinatal metabolic dysregulation is a strong but understudied risk factor for ROP and other long-term sequelae of preterm birth. We will discuss the metabolic and oxygen needs of retina, current treatments, and potential interventions to promote normal vessel growth including control of postnatal hyperglycemia, dyslipidemia and hyperoxia-induced retinal metabolic alterations. Early supplementation of missing nutrients and growth factors and control of supplemental oxygen promotes physiological retinal development. We will discuss the current knowledge gap in retinal metabolism after preterm birth.


Assuntos
Nascimento Prematuro , Neovascularização Retiniana , Retinopatia da Prematuridade , Animais , Gravidez , Feminino , Recém-Nascido , Humanos , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/patologia , Retinopatia da Prematuridade/terapia , Neovascularização Retiniana/metabolismo , Modelos Animais de Doenças , Oxigênio/metabolismo , Fatores de Risco
15.
J Control Release ; 350: 789-802, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35961472

RESUMO

Retinopathy of prematurity (ROP) is characterized by pathological angiogenesis and associated inflammation in the retina and is the leading cause of childhood blindness. MiRNA-223 (miR-223) drives microglial polarization toward the anti-inflammatory phenotype and offers a therapeutic approach to suppress inflammation and consequently pathological neovascularization. However, miRNA-based therapy is hindered by the low stability and non-specific cell-targeting ability of delivery systems. In the present study, we developed folic acid-chitosan (FA-CS)-modified mesoporous silica nanoparticles (PMSN) loaded with miR-223 to regulate retinal microglial polarization. The FA-CS/PMSN/miR-223 nanoparticles exhibited high stability and loading efficiency, achieved targeted delivery, and successfully escaped from lysosomes. In cultured microglial cells, treatment with FA-CS/PMSN/miR-223 nanoparticles upregulated the anti-inflammatory gene YM1/2 and IL-4RA, and downregulated the proinflammatory genes iNOS, IL-1ß, and IL-6. Notably, in a mouse oxygen-induced retinopathy model of ROP, intravitreally injected FA-CS/PMSN/miR-223 nanoparticles (1 µg) decreased the retinal neovascular area by 52.6%. This protective effect was associated with the reduced and increased levels of pro-inflammatory (M1) and anti-inflammatory (M2) cytokines, respectively. Collectively, these findings demonstrate that FA-CS/PMSN/miR-223 nanoparticles provide an effective therapeutic strategy for the treatment of ROP by modulating the miR-223-mediated microglial polarization to the M2 phenotype.


Assuntos
Quitosana , MicroRNAs , Retinopatia da Prematuridade , Animais , Quitosana/uso terapêutico , Modelos Animais de Doenças , Ácido Fólico , Humanos , Imunomodulação , Recém-Nascido , Inflamação , Interleucina-6 , Camundongos , MicroRNAs/genética , MicroRNAs/uso terapêutico , Neovascularização Patológica , Oxigênio/uso terapêutico , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Dióxido de Silício/uso terapêutico
16.
Cell Death Dis ; 13(8): 745, 2022 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-36038541

RESUMO

Current therapies for treatment of proliferative retinopathy focus on retinal neovascularization (RNV) during advanced disease and can trigger adverse side-effects. Here, we have tested a new strategy for limiting neurovascular injury and promoting repair during early-stage disease. We have recently shown that treatment with a stable, pegylated drug form of the ureohydrolase enzyme arginase 1 (A1) provides neuroprotection in acute models of ischemia/reperfusion injury, optic nerve crush, and ischemic stroke. Now, we have determined the effects of this treatment on RNV, vascular repair, and retinal function in the mouse oxygen-induced retinopathy (OIR) model of retinopathy of prematurity (ROP). Our studies in the OIR model show that treatment with pegylated A1 (PEG-A1), inhibits pathological RNV, promotes angiogenic repair, and improves retinal function by a mechanism involving decreased expression of TNF, iNOS, and VEGF and increased expression of FGF2 and A1. We further show that A1 is expressed in myeloid cells and areas of RNV in retinal sections from mice with OIR and human diabetic retinopathy (DR) patients and in blood samples from ROP patients. Moreover, studies using knockout mice with hemizygous deletion of A1 show worsened RNV and retinal injury, supporting the protective role of A1 in limiting the OIR-induced pathology. Collectively, A1 is critically involved in reparative angiogenesis and neuroprotection in OIR. Pegylated A1 may offer a novel therapy for limiting retinal injury and promoting repair during proliferative retinopathy.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Animais , Arginase/genética , Arginase/metabolismo , Modelos Animais de Doenças , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica , Oxigênio , Polietilenoglicóis/uso terapêutico , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/patologia
17.
Int J Mol Sci ; 23(12)2022 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-35742898

RESUMO

Retinopathy of prematurity (ROP) is a severe eye disease leading to blindness. Abnormal vessel formation is the pathological hallmark of neovascular ROP. In forming vessels, vascular endothelial growth factor (VEGF) is an important stimulator. The current anti-ROP therapy has focused on bevacizumab, a monoclonal antibody against VEGF, and pazopanib, a tyrosine kinase inhibitor on the VEGF receptor (VEGFR). Several lines of evidence have proposed that natural compounds may be more effective and safer for anti-VEGF function. Resveratrol, a common natural compound, binds to VEGF and blocks its interaction with VEGFR, thereafter suppressing angiogenesis. Here, we evaluate the efficacy of intravitreal injection, or topical instillation (eye drops), of resveratrol into the eyes of mice suffering from oxygen-induced retinopathy, i.e., developing ROP. The treatment of resveratrol significantly relieved the degree of vascular distortion, permeability and hyperplasia; the efficacy could be revealed by both methods of resveratrol application. In parallel, the treatments of resveratrol inhibited the retinal expressions of VEGF, VEGFR and CD31. Moreover, the applied resveratrol significantly relieved the damage caused by oxygen radicals through upregulating the level of superoxide dismutase (SOD) and downregulating the level of malondialdehyde (MDA) in the retina. Taken together, the potential therapeutic benefit of resveratrol in pro-angiogenic diseases, including retinopathy, can be considered.


Assuntos
Retinopatia da Prematuridade , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Bevacizumab/uso terapêutico , Camundongos , Neovascularização Patológica/tratamento farmacológico , Resveratrol/farmacologia , Resveratrol/uso terapêutico , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/patologia , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
18.
Semin Ophthalmol ; 37(6): 740-748, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35671203

RESUMO

PURPOSE: To evaluate the fluorescein angiography (FA) findings in eyes with spontaneously regressed retinopathy of prematurity (ROP). METHODS: Fluorescein angiography images of 162 eyes of 81 treatment-naive infants who underwent FA due to retinal vascular immaturity and persistent avascular retina (PAR) despite exceeding postmenstrual age of 60 weeks were analyzed retrospectively. Disc diameter (DD), optic disc-to-fovea distance (FD), the length of temporal retinal vascularization (LTRV), and the length of measurable temporal avascular retina distance (LMTAR), were quantitatively measured. RESULTS: The mean gestational age and FA imaging age were 29.39 ± 3.13 and 86.51 ± 24.80 weeks postmenstrual, respectively. The mean ratios of LTRV/FD and LMTAR /DD were 4.47 ± 0.36 and 2.21 ± 1.01, respectively. Pigmentary changes were detected in the peripheral retina in 21% of the eyes. There was at least one angiographic finding in 88% of the eyes, but these findings were usually mild. Based on the FA findings, laser photocoagulation was performed to the peripheral avascular retina in ten eyes of five patients. CONCLUSION: Even in larger preterm infants and without severe retinopathy and anti-VEGF treatment, PAR and peripheral pigmentary changes may be detected, and mild angiographic vascular activity may continue. These findings may lead to late-onset retinal pathologies that may threaten vision. In eyes with PAR, follow-up with FA and prophylactic laser application may be applicable. Further investigation is required for this topic.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Pré-Escolar , Angiofluoresceinografia , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Retina/patologia , Neovascularização Retiniana/diagnóstico , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade/diagnóstico , Retinopatia da Prematuridade/patologia , Estudos Retrospectivos
19.
Adv Sci (Weinh) ; 9(21): e2105365, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35619548

RESUMO

Retinopathy of prematurity (ROP) is one of the leading causes of childhood visual impairment and blindness. However, there are still very few effective pharmacological interventions for ROP. Histone deacetylase 6 (HDAC6)-mediated disassembly of photoreceptor cilia has recently been implicated as an early event in the pathogenesis of ROP. Herein it is shown that enhanced expression of HDAC6 by intravitreal injection of adenoviruses encoding HDAC6 induces the typical pathological changes associated with ROP in mice, including disruption of the membranous disks of photoreceptor outer segments and a decrease in electroretinographic amplitudes. Hdac6 transgenic mice exhibit similar ROP-related defects in retinal structures and functions and disassembly of photoreceptor cilia, whereas Hdac6 knockout mice are resistant to oxygen change-induced retinal defects. It is further shown that blocking HDAC6-mediated cilium disassembly by intravitreal injection of small-molecule compounds protect mice from ROP-associated retinal defects. The findings indicate that pharmacological targeting of the HDAC6-cilium axis may represent a promising strategy for the prevention of ROP.


Assuntos
Cílios , Desacetilase 6 de Histona , Retinopatia da Prematuridade , Animais , Cílios/metabolismo , Cílios/patologia , Desacetilase 6 de Histona/metabolismo , Camundongos , Oxigênio/metabolismo , Retina/metabolismo , Retina/patologia , Retinopatia da Prematuridade/metabolismo , Retinopatia da Prematuridade/patologia
20.
PLoS One ; 17(4): e0267576, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35476813

RESUMO

BACKGROUND: Retinopathy of prematurity (ROP) remains the leading cause for blindness in children. Limited hyperoxia induced proliferative retinopathy (L-HIPR) was recently introduced as a potential animal model for ROP and persistent fetal vasculature; however, the detailed pathological changes remain unclear. METHODS: To model L-HIPR, we placed C57BL/6J mice in 65% oxygen from birth to post-natal day 7 (P7). We examined eyes at intervals between P12 and P30. Retinal morphometry, thickness, and preretinal fibrosis were quantified at different time points on histological sections stained with hematoxylin and eosin (H&E) and Masson Trichrome, respectively. Vascular development, angiogenesis, inflammation, and pericyte coverage were analyzed using immunohistochemistry staining in retinal flat mounts and cross sections. RESULTS: In L-HIPR, the hyaloidal vessels persisted until the latest time point in this study, P30 and began to invaginate the peripheral then central retina starting at P12. Central retinal distortion was noted beginning at P17, while the peripheral retina demonstrated a trend of thinning from P12 to P30. We found that L-HIPR was associated with delayed and abnormal retinal vascular development with subsequent retinal inflammation, pericyte loss and preretinal fibrosis. CONCLUSION: Our study presents a detailed analysis of the L-HIPR animal model demonstrating vitreoretinal pathologic changes, preretinal fibrosis and persistent hyaloidal vessels into adulthood. Based on our findings, we suggest that the persistence and peculiar stepwise migration of the hyaloidal vessels into the retina may provide a potential rescue mechanism for inner retinal development that deserves further study.


Assuntos
Membrana Epirretiniana , Hiperóxia , Neovascularização Retiniana , Retinopatia da Prematuridade , Vitreorretinopatia Proliferativa , Adulto , Animais , Modelos Animais de Doenças , Membrana Epirretiniana/patologia , Fibrose , Humanos , Hiperóxia/complicações , Hiperóxia/patologia , Recém-Nascido , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Retina/patologia , Neovascularização Retiniana/etiologia , Neovascularização Retiniana/patologia , Vasos Retinianos/patologia , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/patologia , Vitreorretinopatia Proliferativa/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA