Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
1.
BMC Pediatr ; 24(1): 390, 2024 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-38858617

RESUMO

BACKGROUND: Oculocutaneous albinism (OCA) is a group of autosomal recessive hereditary disorders that affect melanin biosynthesis, resulting in abnormalities in hair, skin, and eyes. Retinopathy of prematurity (ROP) is a proliferative retinopathy mainly observed in premature infants with low birth weight and early gestational age, but it can also affect full-term infants or children with normal weight, particularly in developing countries. The coexistence of ROP and OCA is rare. There is limited documentation regarding treatment approaches, with few studies reporting positive outcomes with laser treatment due to the absence of melanin pigment. This study discusses the treatment challenges in a female infant diagnosed with ROP and OCA, and underscores the importance of genetic analysis in guiding therapeutic decisions for this rare comorbid condition. CASE PRESENTATION: The study presents a case of ROP occurring concurrently with OCA. Genetic testing revealed two variants, c.727C > T (p.R243C) and c.1832 T > C (p.L611P), in the OCA2 gene, inherited from the patient's mother and father, respectively. The identified mutations were consistent with a diagnosis of OCA2, classified as a subtype of OCA. The patient initially received intravitreal anti-vascular endothelial growth factor (anti-VEGF) injection, followed by laser photocoagulation therapy for a recurrent event. A favorable outcome was observed during the 2-month follow-up period. CONCLUSIONS: The co-occurrence of ROP and OCA is a rare phenomenon, and this is the first recorded case in the Chinese population. The current case supports the use of laser as the primary treatment modality for ROP in OCA2 patients with partial pigmentation impairment. Furthermore, genetic analysis can aid in predicting the effectiveness of laser photocoagulation in this patient population.


Assuntos
Albinismo Oculocutâneo , Retinopatia da Prematuridade , Humanos , Feminino , Albinismo Oculocutâneo/genética , Albinismo Oculocutâneo/complicações , Albinismo Oculocutâneo/terapia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/terapia , Retinopatia da Prematuridade/complicações , Recém-Nascido , Proteínas de Membrana Transportadoras/genética , Mutação , Inibidores da Angiogênese/uso terapêutico , Fotocoagulação a Laser , Bevacizumab/uso terapêutico
2.
Invest Ophthalmol Vis Sci ; 64(7): 11, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37272765

RESUMO

Purpose: This study investigated the associations between vascular endothelial growth factor (VEGF) polymorphisms and retinopathy of prematurity (ROP) risk. Methods: Infants born prematurely at any time from 2009 to 2018 were included. Five single-nucleotide polymorphisms (SNPs) of VEGF were analyzed using real-time PCR in all infants. Multivariate logistic regression was applied to model the associations between VEGF polymorphisms and ROP susceptibility, severity, and premature clinicopathologic characteristics. Results: A total of 334 patients were included and categorized into three groups: those without ROP, those with mild ROP (i.e., ROP not requiring treatment), and those with severe ROP (i.e., ROP for whom treatment was indicated). Among the female patients with ROP, those with VEGF rs3025035 CT (3.231-fold; 95% confidence interval [CI], 1.238-8.431) and a combination of CT and TT genotypes (2.643-fold; 95% CI, 1.056-6.619) exhibited significantly higher risks of severe ROP compared with those with wild-type genotypes. Female ROP infants with VEGF rs3025010 C (TC + CC) alleles had a lower risk of ROP stage ≥3 (odds ratio [OR] = 0.406; 95% CI, 0.165-0.999) than those with TT homozygotes. ROP patients with the VEGF rs10434 A allele (GA + AA) exhibited higher risks of necrotizing enterocolitis (OR = 2.750; 95% CI, 1.119-6.759) and lower risk of bronchopulmonary dysplasia (OR = 0.390; 95% CI, 0.173-0.877) than those with GG homozygotes did. Conclusions: VEGF polymorphisms affect ROP risks differently in male and female infants. In female infants, VEGF rs3025035 with T alleles may predict ROP severity, and VEGF rs3025010 with C alleles may protect against severe ROP.


Assuntos
Retinopatia da Prematuridade , Fator A de Crescimento do Endotélio Vascular , Humanos , Recém-Nascido , Nascimento Prematuro , Fator A de Crescimento do Endotélio Vascular/genética , Retinopatia da Prematuridade/genética
3.
Sci Rep ; 13(1): 143, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36599874

RESUMO

Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are among the most common morbidities affecting extremely premature infants who receive oxygen therapy. Many clinical studies indicate that BPD is associated with advanced ROP. However, the mechanistic link between hyperoxia, BPD, and ROP remains to be explored. Gasdermin D (GSDMD) is a key executor of inflammasome-induced pyroptosis and inflammation. Inhibition of GSDMD has been shown to attenuate hyperoxia-induced BPD and brain injury in neonatal mice. The objective of this study was to further define the mechanistic roles of GSDMD in the pathogenesis of hyperoxia-induced BPD and ROP in mouse models. Here we show that global GSDMD knockout (GSDMD-KO) protects against hyperoxia-induced BPD by reducing macrophage infiltration, improving alveolarization and vascular development, and decreasing cell death. In addition, GSDMD deficiency prevented hyperoxia-induced ROP by reducing vasoobliteration and neovascularization, improving thinning of multiple retinal tissue layers, and decreasing microglial activation. RNA sequencing analyses of lungs and retinas showed that similar genes, including those from inflammatory, cell death, tissue remodeling, and tissue and vascular developmental signaling pathways, were induced by hyperoxia and impacted by GSDMD-KO in both models. These data highlight the importance of GSDMD in the pathogenesis of BPD and ROP and suggest that targeting GSDMD may be beneficial in preventing and treating BPD and ROP in premature infants.


Assuntos
Displasia Broncopulmonar , Gasderminas , Retinopatia da Prematuridade , Animais , Camundongos , Animais Recém-Nascidos , Displasia Broncopulmonar/genética , Displasia Broncopulmonar/metabolismo , Modelos Animais de Doenças , Hiperóxia/complicações , Hiperóxia/metabolismo , Hipertensão Pulmonar/patologia , Pulmão/patologia , Proteínas de Ligação a Fosfato/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/complicações , Gasderminas/genética , Gasderminas/metabolismo
4.
Int J Mol Sci ; 23(13)2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35806359

RESUMO

The pathophysiology of retinopathy of prematurity (ROP) is postulated to first involve delayed intraretinal vascularization, followed by intravitreal neovascularization (IVNV). Although intravitreal agents that reduce the bioactivity of vascular endothelial growth factor (VEGF) are used to treat IVNV, concerns exist regarding their effects on intraretinal vascularization. In an experimental ROP model, VEGF receptor 2 (VEGFR2) knockdown in retinal endothelial cells reduced IVNV and promoted intraretinal vascularization, whereas knockdown of a downstream effector, signal transducer and activator of transcription 3 (STAT3) in retinal endothelial cells only reduced IVNV. In this study, we tested the hypothesis that the different pathways involved in VEGF-triggered VEGFR2 signaling and VEGF-triggered STAT3 signaling in retinal endothelial cells would allow us to delineate signaling pathways involved in IVNV from those involved in intraretinal vascularization in ROP. To address our hypothesis, we used RNA-sequencing and pathway enrichment analysis to determine changes in the transcriptome of cultured human retinal microvascular endothelial cells (HRMECs). Of the enriched pathways, inactivation of oncostatin M signaling was predicted by either KDR or STAT3 knockdown in the presence of VEGF. Activation of kinetochore metaphase signaling was predicted by KDR knockdown, whereas inactivation was predicted by STAT3 knockdown in the presence of VEGF. Inactivation of signaling by the Rho family of GTPases was predicted by KDR knockdown, but activation was predicted by STAT3 knockdown in the presence of VEGF. Taken together, our data identified unique signaling pathway differences between VEGF-triggered VEGFR2 and VEGF-triggered STAT3 in HRMECs that might have implications in ROP.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Humanos , Recém-Nascido , Neovascularização Patológica/metabolismo , RNA-Seq , Ratos , Ratos Sprague-Dawley , Neovascularização Retiniana/metabolismo , Vasos Retinianos/metabolismo , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Ophthalmic Genet ; 43(4): 543-549, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35416114

RESUMO

BACKGROUND: Coats plus syndrome or cerebroretinal microangiopathy with calcifications and cysts (CMCC) is an exceedingly rare autosomal recessive disorder that predominantly affects the microvasculature in the retina, brain, bones, and gastrointestinal system. Unlike Coats disease, CMCC is bilateral and affects multiple organ systems. MATERIALS AND METHODS: Case report. RESULTS: We report the case of two brothers with Coats Plus syndrome who presented with variable phenotypic expression. One sibling (Patient 1) was thought to have atypical retinopathy of prematurity and was only diagnosed with Coats plus after his older brother (Patient 2) presented with a seizure and a left upper extremity tremor at 4 years of age. The CTC1 mutation was confirmed in both patients. Aggressive treatment with laser photocoagulation and intravitreal bevacizumab dramatically improved the retinal vascular and exudative changes. CONCLUSION: Coats Plus syndrome can have a variable phenotypic presentation, including retinal vascular findings. This rare genetic disease should be in the differential diagnosis in patients who present with atypical retinal pathology, including Retinopathy of Prematurity, Familial Exudative Vitreoretinopathy, or Coats disease associated with non-specific multiorgan abnormalities.


Assuntos
Cistos do Sistema Nervoso Central , Leucoencefalopatias , Telangiectasia Retiniana , Retinopatia da Prematuridade , Ataxia , Neoplasias Encefálicas , Calcinose , Cistos do Sistema Nervoso Central/genética , Humanos , Recém-Nascido , Fotocoagulação a Laser , Leucoencefalopatias/genética , Masculino , Espasticidade Muscular , Doenças Retinianas , Telangiectasia Retiniana/diagnóstico , Telangiectasia Retiniana/genética , Telangiectasia Retiniana/terapia , Retinopatia da Prematuridade/diagnóstico , Retinopatia da Prematuridade/genética , Convulsões
6.
Mol Cell Biochem ; 477(6): 1739-1763, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35262882

RESUMO

Retinopathy of prematurity (ROP) is a retinal vasoproliferative disorder that represents an important cause of childhood visual impairment and blindness. Although oxidative stress has long been implicated in ROP etiology, other prenatal and perinatal factors are also involved. This review focuses on current research involving inflammation and genetic factors in the pathogenesis of ROP. Increasing evidence suggests that perinatal inflammation or infection contributes to ROP pathogenesis. Cytokines and chemokines with a fundamental role in inflammatory responses and that significantly contributing to angiogenesis are analyzed. Microglia cells, the retinal-resident macrophages, are crucial for retinal homeostasis, however, under sustained pathological stimuli release exaggerated amounts of inflammatory mediators and can promote pathological neovascularization. Current modulation of angiogenic cytokines, such as treatment with antibodies to vascular endothelial growth factor (anti-VEGF), has shown efficacy in the treatment of ocular neovascularization; however, some patients are refractory to anti-VEGF agents, suggesting that other angiogenic or anti-angiogenic cytokines need to be identified. Much evidence suggests that genetic factors contribute to the phenotypic variability of ROP. Several studies have implicated the involvement of candidate genes from different signaling pathways in the development of ROP. However, a genetic component with a major impact on ROP has not yet been discovered. Most studies have limitations and did not replicate results. Future research involving bioinformatics, genomics, and proteomics may contribute to finding more genes associated with ROP and may allow discovering better solutions in the management and treatment of ROP.


Assuntos
Retinopatia da Prematuridade , Citocinas/genética , Humanos , Recém-Nascido , Inflamação/genética , Neovascularização Patológica/genética , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Fator A de Crescimento do Endotélio Vascular/genética
7.
J Matern Fetal Neonatal Med ; 35(7): 1248-1257, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32264735

RESUMO

PURPOSE: To investigate the influence of polymorphisms in vascular endothelial growth factor (VEGF), tumor necrosis factor-α (TNF-α), and glutathione S-transferase Pi isoform (GSTP1) genes on retinopathy of prematurity (ROP) risk, we performed a Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)-compliant meta-analysis. METHODS: An exhaustive search was conducted in PubMed, Web of Science, and CNKI for genetic studies evaluating the relationship between VEGF (-460 T/C, +936 C/T, -634 G/C, and -2578 C/A), TNF-α (-308 G/A) and GSTP1 (Ile/Val) polymorphisms and ROP risk from inception until November 2019. Odds ratio (OR) with the 95% confidence interval (CI) were used for estimating combined effect size. The quality of the included studies was evaluated using the Newcastle-Ottawa Scale (NOS). RESULTS: A total of 14 studies met the inclusion criteria. The meta-analyses revealed that VEGF - 460 T/C was associated with ROP risk in the allele model (C vs. T, OR = 0.83, 95% CI: 0.74-0.94, POR=0.004), homozygous gene model (CC vs. TT, OR = 0.70, 95% CI: 0.54-0.91, POR=0.008), dominant gene model (CC + TC vs. TT, OR = 0.80, 95% CI: 0.67-0.95, POR = 0.012), and recessive gene model (CC vs. TC + TT, OR = 0.74, 95% CI: 0.59-0.94, POR = 0.014). However, we did not find significant differences in the genotype and allele distribution of VEGF + 936 C/T, -634 G/C, -2578 C/A, TNF-α - 308 G/A and GSTP1 Ile/Val polymorphisms, between ROP and control group (p > .05). CONCLUSIONS: VEGF polymorphism -460 T/C was associated with a lower ROP risk. Further research is warranted to investigate haplotype effects of VEGF polymorphisms on the risk of ROP.


Assuntos
Retinopatia da Prematuridade , Fator de Necrose Tumoral alfa , Alelos , Predisposição Genética para Doença , Glutationa S-Transferase pi/genética , Humanos , Recém-Nascido , Polimorfismo de Nucleotídeo Único , Retinopatia da Prematuridade/genética , Fatores de Risco , Fator de Necrose Tumoral alfa/genética , Fator A de Crescimento do Endotélio Vascular/genética
8.
Pediatr Res ; 91(7): 1677-1685, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34285351

RESUMO

BACKGROUND: Pathologic ocular neovascularization in retinopathy of prematurity (ROP) and other proliferative retinopathies are characterized by dysregulation of vascular endothelial growth factor-A (VEGF-A). A study of Vegfa isoform expression during oxygen-induced ischemic retinopathy (OIR) may enhance our understanding of Vegf dysregulation. METHODS: Following induction of OIR, immunohistochemistry and polymerase chain reaction (PCR) was performed on room air (RA) and OIR mice. RESULTS: Total Vegfa messenger RNA (mRNA) expression was stable in RA mice, but increased in OIR mice with a peak at postnatal day 17 (P17), before returning to RA levels. Vegfa164a expression was similar in both OIR and RA mice at P10 (Phase 1 OIR), but 2.4-fold higher in OIR mice compared to RA mice at P16 (Phase 2 OIR). At P10, Vegfa164b mRNA was similar in OIR vs RA mice, but was expressed 2.5-fold higher in OIR mice compared to RA mice at P16. At P10 and P16, Vegfr2/Vegfr1 expression was increased in OIR mice compared to RA mice. Increased activation of microglia was seen in OIR mice. CONCLUSIONS: Vegfa164a, Vegfa164b, and Vegfr1 were overexpressed in OIR mice, leading to abnormal signaling and angiogenesis. Further studies of mechanisms of Vegf dysregulation may lead to novel therapies for ROP and other proliferative retinopathies. IMPACT: Vegfa164 has two major isoforms, a proangiogenic, Vegfa164a, and an antiangiogenic, Vegfa164b, with opposing receptors, inhibitory Vegfr1, and stimulatory Vegfr2, but their role in OIR is unclear. In Phase 1 OIR, both isoforms and receptors are expressed similarly. In Phase 2 OIR, both isoforms are overexpressed, with an increased ratio of inhibitory Vegfr1. Modulation of angiogenesis by Vegf regulation enables pruning of excess angiogenesis during physiology, but results in ineffective angiogenesis during OIR. Knowledge of VEGF dysregulation may have novel therapeutic implications in the management of ROP and retinal proliferative diseases.


Assuntos
Neovascularização Retiniana , Retinopatia da Prematuridade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Patológica/metabolismo , Oxigênio/uso terapêutico , Isoformas de Proteínas , RNA Mensageiro/metabolismo , Neovascularização Retiniana/genética , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia
9.
Ophthalmic Genet ; 42(6): 725-731, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34346275

RESUMO

BACKGROUND: In addition to risk factors such as low birth weight and uncontrolled oxygen therapy, genetic predisposition is also thought to play a role in the development of retinopathy of prematurity (ROP). In our study, we aimed to analyze single-nucleotide polymorphisms (SNPs) in VEGFA, EPAS1, BDNF and NOS3 genes in infants who develop ROP. MATERIALS AND METHODS: Seventy-five mild-moderate and 73 severe ROP cases were included in this study. Eleven different SNPs regions that located in VEGFA, EPAS1, BDNF and NOS3 genes were analysed by SnapShot technique and compared between two groups by the multiple logistic regression analysis. RESULTS: Statistically significant results were obtained in 8 of the 11 SNPs. It was observed that the excess of mutant alleles in four (VEGFA rs2010963 and rs3025039, EPAS1 rs13419896, NOS3 rs2070744) of these regions increased ROP severity and treatment requirement (p < .001, p < .001, p = .022, p = .004, respectively) while the excess of mutant alleles in the other four regions (VEGFA rs833061, BDNF rs7929344, EPAS1 rs1867785 and rs1868085) showed that ROP severtiy was milder and eliminated the need for treatment (p < .001, p = .019, p = .017, p = .017, respectively). CONCLUSIONS: Considering the results of our study, it was seen that besides the known environmental and demographic factors in ROP pathogenesis, genetic predisposition also had an effect on the clinic and course of ROP. Polymorphisms of VEGFA rs2010963 and rs3025039, EPAS1 rs13419896, NOS3 rs2070744 were found to be associated with severe ROP. More studies involving different populations cases are needed to confirm these findings and enlighten the etiology of ROP.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fator Neurotrófico Derivado do Encéfalo/genética , Óxido Nítrico Sintase Tipo III/genética , Polimorfismo de Nucleotídeo Único/genética , Retinopatia da Prematuridade/genética , Fator A de Crescimento do Endotélio Vascular/genética , Alelos , Feminino , Seguimentos , Frequência do Gene , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Masculino , Retinopatia da Prematuridade/diagnóstico , Fatores de Risco
10.
Exp Eye Res ; 210: 108716, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34352266

RESUMO

PURPOSE: To evaluate the role of Toll-like receptor 2 (TLR2) signaling in retinal neovascularization in a mouse model of oxygen-induced retinopathy (OIR). MATERIALS AND METHODS: The OIR model was established in C57BL/6J wild type (WT) mice and TLR2-/- mice. Retinal neovascularization in the OIR model was measured by counting new vascular cell nuclei above the internal limiting membrane and analyzing flat-mounted retinas perfused with fluorescein dextran and immunostained with Griffonia Simplicifolia (GS) isolectin. The expression of TLR2 and VEGF in the retina was detected by immunofluorescence. Expression of TGF- ß1, b-FGF, and IL-6 mRNA in the retina was measured by quantitative real-time PCR. RESULTS: Compared to WT OIR mice, retinal neovascularization was attenuated in TLR2-/- OIR mice. The co-expressions of TLR2 and VEGF were remarkably and consistently increased in WT OIR mice; however, there was no expression of TLR2 and a significant decrease in VEGF expression in TLR2-/- OIR mice. These results suggest that TLR2 plays a central role in OIR model angiogenesis. Expression of TGF- ß1, b-FGF, and IL-6 mRNA were reduced in the TLR2-/- OIR mice, suggesting that the inflammatory response induced by TLR2 relates to angiogenesis. CONCLUSION: TLR2 signaling in the retina is associated with neovascularization in mice. Inflammation contributes to the activation of angiogenesis and is partially mediated through the TLR2-VEGF retinal signaling pathway.


Assuntos
Modelos Animais de Doenças , Oxigênio/toxicidade , Neovascularização Retiniana/metabolismo , Retinopatia da Prematuridade/metabolismo , Transdução de Sinais/fisiologia , Receptor 2 Toll-Like/metabolismo , Animais , Citocinas/genética , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Neovascularização Retiniana/genética , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Receptor 2 Toll-Like/genética , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Int J Mol Sci ; 22(9)2021 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34062733

RESUMO

Retinopathy of prematurity (ROP) is an ocular vascular disease affecting premature infants, characterized by pathological retinal neovascularization (RNV), dilated and tortuous retinal blood vessels, and retinal or vitreous hemorrhages that may lead to retinal detachment, vision impairment and blindness. Compared with other neovascular diseases, ROP is unique because of ongoing and concurrent physiological and pathological angiogenesis in the developing retina. While the disease is currently treated by laser or cryotherapy, anti-vascular endothelial growth factor (VEGF) agents have been extensively investigated but are not approved in the U.S. because of safety concerns that they negatively interfere with physiological angiogenesis of the developing retina. An ideal therapeutic strategy would selectively inhibit pathological but not physiological angiogenesis. Our group recently described a novel strategy that selectively and safely alleviates pathological RNV in animal models of ROP by targeting secretogranin III (Scg3), a disease-restricted angiogenic factor. The preclinical profile of anti-Scg3 therapy presents a high potential for next-generation disease-targeted anti-angiogenic therapy for the ROP indication. This review focuses on retinal vessel development in neonates, the pathogenesis of ROP and its underlying molecular mechanisms, including different animal models, and provides a summary of current and emerging therapies.


Assuntos
Cromograninas/genética , Neovascularização Patológica/tratamento farmacológico , Oxigênio/uso terapêutico , Retinopatia da Prematuridade/tratamento farmacológico , Animais , Animais Recém-Nascidos , Cromograninas/antagonistas & inibidores , Humanos , Camundongos , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/genética , Retina/efeitos dos fármacos , Retina/crescimento & desenvolvimento , Retina/patologia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Fator A de Crescimento do Endotélio Vascular/genética
12.
J Mol Neurosci ; 71(12): 2546-2557, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33895966

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide having trophic and protective functions in neural tissues, including the retina. Previously, we have shown that intravitreal PACAP administration can maintain retinal structure in the animal model of retinopathy of prematurity (ROP). The purpose of this study is to examine the development of ROP in PACAP-deficient and wild-type mice to reveal the function of endogenous PACAP. Wild-type and PACAP-knockout (KO) mouse pups at postnatal day (PD) 7 were maintained at 75% oxygen for 5 consecutive days then returned to room air on PD12 to develop oxygen-induced retinopathy (OIR). On PD15, animals underwent electroretinography (ERG) to assess visual function. On PD16, eyes were harvested for either immunohistochemistry to determine the percentage of the central avascular retinal area or molecular analysis to assess angiogenesis proteins by array kit and anti-apoptotic protein kinase B (Akt) change by western blot. Retinas of PACAP-deficient OIR mice showed a greater central avascular area than that of the wild types. ERG revealed significantly decreased b-wave amplitude in PACAP KO compared to their controls. Several angiogenic proteins were upregulated due to OIR, and 11 different proteins markedly increased in PACAP-deficient mice, whereas western blot analysis revealed a reduction in Akt phosphorylation, suggesting an advanced cell death in the lack of PACAP. This is the first study to examine the endogenous effect of PACAP in the OIR model. Previously, we have shown the beneficial effect of exogenous local PACAP treatment in the rat OIR model. Together with the present findings, we suggest that PACAP could be a novel retinoprotective agent in ROP.


Assuntos
Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/metabolismo , Retinopatia da Prematuridade/metabolismo , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/toxicidade , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Retina/metabolismo , Retina/fisiopatologia , Retinopatia da Prematuridade/etiologia , Retinopatia da Prematuridade/genética , Visão Ocular
13.
Fetal Pediatr Pathol ; 40(3): 233-249, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31846376

RESUMO

BACKGROUND: Previous studies have suggested an association between VEGF-A polymorphisms and retinopathy of prematurity (ROP) risk. But the conclusions are still controversial. The aim of this meta-analysis was to evaluate the association between the VEGF-A polymorphisms and susceptibility of ROP. Methods: We searched PubMed, Scopus, WanFang and CNKI databases for all eligible case-control studies published before September 30, 2019. Results: A total of 27 case-control studies with 5,748 ROP cases and 6,146 controls were selected. The results suggested that there was an association between VEGF-A -460T > C polymorphism and increased risk of ROP under the allele model (C vs. T: OR= 0.879, 95% CI 0.776-0.994, p = 0.040). However, VEGF-A -634G > C, +405G > C and +936C > T polymorphisms were not significantly associated with risk of ROP. The subgroup analysis demonstrated that VEGF-A +405G > C polymorphism was associated with ROP risk in Caucasians. Conclusions: This meta-analysis indicates that VEGF-A -460T > C polymorphism may contribute to the susceptibility for ROP.


Assuntos
Retinopatia da Prematuridade , Fator A de Crescimento do Endotélio Vascular/genética , Alelos , Estudos de Casos e Controles , Predisposição Genética para Doença , Humanos , Recém-Nascido , Polimorfismo de Nucleotídeo Único , Retinopatia da Prematuridade/genética
14.
Clin Epigenetics ; 12(1): 151, 2020 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-33076993

RESUMO

BACKGROUND: Infants born very preterm are more likely to experience neonatal morbidities compared to their term peers. Variations in DNA methylation (DNAm) associated with these morbidities may yield novel information about the processes impacted by these morbidities. METHODS: This study included 532 infants born < 30 weeks gestation, participating in the Neonatal Neurobehavior and Outcomes in Very Preterm Infants study. We used a neonatal morbidity risk score, which was an additive index of the number of morbidities experienced during the NICU stay, including bronchopulmonary dysplasia (BPD), severe brain injury, serious neonatal infections, and severe retinopathy of prematurity. DNA was collected from buccal cells at discharge from the NICU, and DNAm was measured using the Illumina MethylationEPIC. We tested for differential methylation in association with the neonatal morbidity risk score then tested for differentially methylated regions (DMRs) and overrepresentation of biological pathways. RESULTS: We identified ten differentially methylated CpGs (α Bonferroni-adjusted for 706,278 tests) that were associated with increasing neonatal morbidity risk scores at three intergenic regions and at HPS4, SRRD, FGFR1OP, TNS3, TMEM266, LRRC3B, ZNF780A, and TENM2. These mostly followed dose-response patterns, for 8 CpGs increasing DNAm associated with increased numbers of morbidities, while for 2 CpGs the risk score was associated with decreasing DNAm. BPD was the most substantial contributor to differential methylation. We also identified seven potential DMRs and over-representation of genes involved in Wnt signaling; however, these results were not significant after Bonferroni adjustment for multiple testing. CONCLUSIONS: Neonatal DNAm, within genes involved in fibroblast growth factor activities, cellular invasion and migration, and neuronal signaling and development, are sensitive to the neonatal health complications of prematurity. We hypothesize that these epigenetic features may be representative of an integrated marker of neonatal health and development and are promising candidates to integrate with clinical information for studying developmental impairments in childhood.


Assuntos
Metilação de DNA/genética , Epigenômica/métodos , Doenças do Prematuro/genética , Recém-Nascido Prematuro/metabolismo , Morbidade/tendências , Adulto , Lesões Encefálicas/diagnóstico , Lesões Encefálicas/genética , Displasia Broncopulmonar/diagnóstico , Displasia Broncopulmonar/genética , Ilhas de CpG/genética , Feminino , Idade Gestacional , Humanos , Recém-Nascido , Doenças do Prematuro/etnologia , Infecções/diagnóstico , Infecções/genética , Masculino , Mucosa Bucal/metabolismo , Gravidez , Retinopatia da Prematuridade/diagnóstico , Retinopatia da Prematuridade/genética , Fatores de Risco , Índice de Gravidade de Doença
15.
Biomed Pharmacother ; 132: 110856, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33125970

RESUMO

PURPOSE: The incidence of retinopathy of prematurity (ROP) has increased continuously in recent years. However, the therapeutic effects of current treatments still remain undesired. This study aims to investigate the role of C-CBL in retinal angiogenesis in ROP and its potential as a therapeutic target. METHODS: Mouse retina microvascular endothelial cells (mRMECs) and induced experimental ROP/ oxygen-induced retinopathy (OIR) mice were employed to investigate the role of C-CBL in angiogenesis with combined molecular and cellular approaches, and histopathology methods. OIR mouse pups at postnatal day 12 (P12) were either injected intravitreally with adenovirus overexpressing c-Cbl or c-Cbl siRNA. Retinal neovascularization and avascular status were evaluated by retinal immunofluorescence (IF) staining, whole-mounts and hematoxylin and eosin (H&E) staining. RESULTS: C-CBL inhibits neovascularization by negatively regulating JAK2/STAT3/VEGF signaling axis in a ubiquitination-dependent manner. Knockdown of c-Cbl by siRNA reduced ubiquitin-mediated JAK2 degradation and increased levels of p-JAK2, p-STAT3, VEGF, and neovascularization in mRMECs, which can be reversed by JAK2 inhibitor treatment. While knockdown of c-Cbl significantly increased neovascular (NV) zone in the retinas, c-Cbl overexpression inhibited neovascularization in the retinal tissues in OIR mice. CONCLUSION: We found that C-CBL is required for anti-neovascularization process in ROP development by inhibiting JAK2/STAT3-dependent angiogenesis. Thus, our finding strongly suggest that C-CBL may be a potential novel therapeutic target for treating ROP.


Assuntos
Proteínas Proto-Oncogênicas c-cbl/genética , Neovascularização Retiniana/patologia , Retinopatia da Prematuridade/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Técnicas de Silenciamento de Genes , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio , Neovascularização Retiniana/genética , Vasos Retinianos/citologia , Vasos Retinianos/patologia , Retinopatia da Prematuridade/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Exp Mol Med ; 51(10): 1-13, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31604908

RESUMO

Angiogenesis should be precisely regulated because disordered neovascularization is involved in the aggravation of multiple diseases. The vascular endothelial growth factor (VEGF)-A/VEGF receptor 2 (VEGFR-2) axis is crucial for controlling angiogenic responses in vascular endothelial cells (ECs). Therefore, inactivating VEGFR-2 signaling may effectively suppress aberrant angiogenesis and alleviate related symptoms. In this study, we performed virtual screening, identified the synthetic disaccharide 6'-sialylgalactose (6SG) as a potent VEGFR-2-binding compound and verified its high binding affinity by Biacore assay. 6SG effectively suppressed VEGF-A-induced VEGFR-2 phosphorylation and subsequent in vitro angiogenesis in HUVECs without inducing cytotoxicity. 6SG also inhibited VEGF-A-induced extracellular-regulated kinase (ERK)/Akt activation and actin stress fiber formation in HUVECs. We demonstrated that 6SG inhibited retinal angiogenesis in a mouse model of retinopathy of prematurity and tumor angiogenesis in a xenograft mouse model. Our results suggest a potential therapeutic benefit of 6SG in inhibiting angiogenesis in proangiogenic diseases, such as retinopathy and cancer.


Assuntos
Galactose/metabolismo , Neoplasias/genética , Neovascularização Patológica/genética , Fator A de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Inibidores da Angiogênese/metabolismo , Animais , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Galactose/análogos & derivados , Xenoenxertos , Células Endoteliais da Veia Umbilical Humana , Humanos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Fosforilação/genética , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Transdução de Sinais/genética , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
17.
Cell Cycle ; 18(22): 3147-3159, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31564208

RESUMO

Since the functions of Astragalus root extract in retinopathy remain to be unraveled, this study is performed to elucidate whether Astragalus root extract functions in retinal cell apoptosis and angiogenesis in retinopathy of prematurity (ROP). Newborn mice were selected for establishing mice models of oxygen-induced retinopathy (OIR), which were treated with high-, medium- or low-Astragalus root extract. Evans Blue (EB) was perfused to detect the blood retinal barrier. Additionally, the vascular morphology, number of endothelial cell nuclei of neovascularization, proliferation of blood vessels, ultrastructural changes were determined via a series of assays. Moreover, levels of reactive oxygen species (ROS), expression of other factors such as VEGF, PEDF, IGF-1, HIF-1α, Bax, Bcl-2, eNOS, nNOS, and iNOS were detected. Astragalus root extract was found to protect blood-retinal barrier in the OIR model mice through repairing the structure and morphology of retina, inhibiting ROS production, retinal cell apoptosis, as well as improving retinal vascular angiogenesis. Astragalus root extract was also found to decrease VEGF and HIF-1α expression, but enhance PEDF and IGF-1 expression in the OIR model mice, thereby protecting retinas in ROP. This study highlights that Astragalus root extract is able to suppress retinal cell apoptosis and repair damaged retinal neovascularization in ROP, which provides basis for ROP therapy.


Assuntos
Apoptose/efeitos dos fármacos , Astrágalo/química , Neovascularização Patológica/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Retina/efeitos dos fármacos , Vasos Retinianos , Retinopatia da Prematuridade/tratamento farmacológico , Retinopatia da Prematuridade/metabolismo , Animais , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Neovascularização Patológica/metabolismo , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/metabolismo , Óxido Nítrico Sintase Tipo I/genética , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Oxigênio/toxicidade , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Retina/citologia , Retina/patologia , Retina/ultraestrutura , Retinopatia da Prematuridade/induzido quimicamente , Retinopatia da Prematuridade/genética , Serpinas/genética , Serpinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Proteína X Associada a bcl-2/metabolismo
18.
Am J Pathol ; 189(11): 2340-2356, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31430465

RESUMO

Retinopathy of prematurity (ROP) is characterized by an initial retinal avascularization, followed by pathologic neovascularization. Recently, choroidal thinning has also been detected in children formerly diagnosed with ROP; a similar sustained choroidal thinning is observed in ROP models. But the mechanism underlying the lack of choroidal revascularization remains unclear and was investigated in an oxygen-induced retinopathy (OIR) model. In OIR, evidence of senescence was detected, preceded by oxidative stress in the choroid and the retinal pigment epithelium. This was associated with a global reduction of proangiogenic factors, including insulin-like growth factor 1 receptor (Igf1R). Coincidentally, tumor suppressor p53 was highly expressed in the OIR retinae. Curtailing p53 activity resulted in reversal of senescence, normalization of Igf1r expression, and preservation of choroidal integrity. OIR-induced down-regulation of Igf1r was mediated at least partly by miR-let-7b as i) let-7b expression was augmented throughout and beyond the period of oxygen exposure, ii) let-7b directly targeted Igf1r mRNA, and iii) p53 knock-down blunted let-7b expression, restored Igf1r expression, and elicited choroidal revascularization. Finally, restoration of Igf1r expression rescued choroid thickness. Altogether, this study uncovers a significant mechanism for defective choroidal revascularization in OIR, revealing a new role for p53/let-7b/IGF-1R axis in the retina. Future investigations on this (and connected) pathway could further our understanding of other degenerative choroidopathies, such as geographic atrophy.


Assuntos
Corioide/irrigação sanguínea , Corioide/efeitos dos fármacos , MicroRNAs/fisiologia , Neovascularização Fisiológica/efeitos dos fármacos , Oxigênio/efeitos adversos , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Corioide/metabolismo , Corioide/patologia , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Células HEK293 , Humanos , Neovascularização Fisiológica/genética , Oxigênio/farmacologia , Ratos , Ratos Long-Evans , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Retinopatia da Prematuridade/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
19.
Invest Ophthalmol Vis Sci ; 60(8): 2888-2894, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31266060

RESUMO

Purpose: Extremely preterm infants are at increased risk for retinopathy of prematurity (ROP). We previously identified several inflammatory proteins that were expressed early in life and are associated with an increased risk of ROP and several angiogenic and neurotrophic growth factors in the neonatal systemic circulation that are associated with a lower risk of ROP. In this paper, we report the results of a set of analyses designed to test the hypothesis that placental CpG methylation levels of 12 inflammation-, angiogenic-, and neurotrophic-associated genes predict the occurrence of prethreshold ROP in extremely preterm newborns. Methods: We used placental CpG methylation data from 395 newborns from the Extremely Low Gestational Age Newborns study. Results: Multivariable regression models revealed that placental DNA methylation of 16 CpG sites representing 8 genes were associated with prethreshold ROP. Specifically, CpG methylation in the serum amyloid A SAA1 and SAA2, brain-derived neurotrophic factor (BDNF), myeloperoxidase (MPO), C-reactive protein (CRP), angiopoietin 1 (ANGPT1), and tumor necrosis factor receptor superfamily member 1B (TNFRSF1B) genes was associated with a lower risk of prethreshold ROP. Conversely, CpG methylation at three probes within tumor necrosis factor receptor superfamily member 1A (TNFRSF1A) and in two alternative probes within the BDNF and ANGPT1 genes was associated with an increased risk of ROP. Conclusions: CpG methylation may be a useful marker for improving ROP prediction, opening the opportunity for early intervention to lessen disease severity.


Assuntos
Proteínas Angiogênicas/genética , Ilhas de CpG/fisiologia , Metilação de DNA , Inflamação/genética , Fatores de Crescimento Neural/genética , Placenta/metabolismo , Retinopatia da Prematuridade/genética , Adulto , Feminino , Idade Gestacional , Humanos , Recém-Nascido de Peso Extremamente Baixo ao Nascer , Lactente Extremamente Prematuro , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Razão de Chances , Gravidez , Fatores de Risco
20.
J Cell Mol Med ; 23(4): 2362-2371, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30680928

RESUMO

The mechanism underlying vasoproliferative retinopathies like retinopathy of prematurity (ROP) is hypoxia-triggered neovascularisation. Nerve growth factor (NGF), a neurotrophin supporting survival and differentiation of neuronal cells may also regulate endothelial cell functions. Here we studied the role of NGF in pathological retinal angiogenesis in the course of the ROP mouse model. Topical application of NGF enhanced while intraocular injections of anti-NGF neutralizing antibody reduced pathological retinal vascularization in mice subjected to the ROP model. The pro-angiogenic effect of NGF in the retina was mediated by inhibition of retinal endothelial cell apoptosis. In vitro, NGF decreased the intrinsic (mitochondria-dependent) apoptosis in hypoxia-treated human retinal microvascular endothelial cells and preserved the mitochondrial membrane potential. The anti-apoptotic effect of NGF was associated with increased BCL2 and reduced BAX, as well as with enhanced ERK and AKT phosphorylation, and was abolished by inhibition of the AKT pathway. Our findings reveal an anti-apoptotic role of NGF in the hypoxic retinal endothelium, which is involved in promoting pathological retinal vascularization, thereby pointing to NGF as a potential target for proliferative retinopathies.


Assuntos
Anticorpos Neutralizantes/farmacologia , Neovascularização Patológica/terapia , Fator de Crescimento Neural/antagonistas & inibidores , Retinopatia da Prematuridade/terapia , Apoptose/efeitos dos fármacos , Células Endoteliais , Humanos , Injeções Intraoculares , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fator de Crescimento Neural/genética , Neurônios/efeitos dos fármacos , Neurônios/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Retina/efeitos dos fármacos , Retina/patologia , Retinopatia da Prematuridade/genética , Retinopatia da Prematuridade/patologia , Proteína X Associada a bcl-2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA