Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 402
Filter
1.
Invest Ophthalmol Vis Sci ; 65(8): 10, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38958972

ABSTRACT

Purpose: Retinopathy of prematurity (ROP) results from postnatal hyperoxia exposure in premature infants and is characterized by aberrant neovascularization of retinal blood vessels. Epithelial membrane protein-2 (EMP2) regulates hypoxia-inducible factor (HIF)-induced vascular endothelial growth factor (VEGF) production in the ARPE-19 cell line and genetic knock-out of Emp2 in a murine oxygen-induced retinopathy (OIR) model attenuates neovascularization. We hypothesize that EMP2 blockade via intravitreal injection protects against neovascularization. Methods: Ex vivo choroid sprouting assay was performed, comparing media and human IgG controls versus anti-EMP2 antibody (Ab) treatment. In vivo, eyes from wild-type (WT) mice exposed to hyperoxia from postnatal (P) days 7 to 12 were treated with P12 intravitreal injections of control IgG or anti-EMP2 Abs. Neovascularization was assessed at P17 by flat mount imaging. Local and systemic effects of anti-EMP2 Ab treatment were assessed. Results: Choroid sprouts treated with 30 µg/mL of anti-EMP2 Ab demonstrated a 48% reduction in vessel growth compared to control IgG-treated sprouts. Compared to IgG-treated controls, WT OIR mice treated with 4 µg/g of intravitreal anti-EMP2 Ab demonstrated a 42% reduction in neovascularization. They demonstrated down-regulation of retinal gene expression in pathways related to vasculature development and up-regulation in genes related to fatty acid oxidation and tricarboxylic acid cycle respiratory electron transport, compared to controls. Anti-EMP2 Ab-treated OIR mice did not exhibit gross retinal histologic abnormalities, vision transduction abnormalities, or weight loss. Conclusions: Our results suggest that EMP2 blockade could be a local and specific treatment modality for retinal neovascularization in oxygen-induced retinopathies, without systemic adverse effects.


Subject(s)
Animals, Newborn , Disease Models, Animal , Intravitreal Injections , Mice, Inbred C57BL , Oxygen , Retinal Neovascularization , Retinopathy of Prematurity , Animals , Mice , Oxygen/toxicity , Retinal Neovascularization/metabolism , Retinal Neovascularization/prevention & control , Retinal Neovascularization/pathology , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Hyperoxia/complications , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Humans
2.
Transl Vis Sci Technol ; 13(5): 1, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691083

ABSTRACT

Purpose: This feasibility study investigated the practicability of collecting and analyzing tear proteins from preterm infants at risk of retinopathy of prematurity (ROP). We sought to identify any tear proteins which might be implicated in the pathophysiology of ROP as well as prognostic markers. Methods: Schirmer's test was used to obtain tear samples from premature babies, scheduled for ROP screening, after parental informed consent. Mass spectrometry was used for proteomic analysis. Results: Samples were collected from 12 infants, which were all adequate for protein analysis. Gestational age ranged from 25 + 6 to 31 + 1 weeks. Postnatal age at sampling ranged from 19 to 66 days. One infant developed self-limiting ROP. Seven hundred one proteins were identified; 261 proteins identified in the majority of tear samples, including several common tear proteins, were used for analyses. Increased risk of ROP as determined by the postnatal growth ROP (G-ROP) criteria was associated with an increase in lactate dehydrogenase B chain in tears. Older infants demonstrated increased concentration of immunoglobulin complexes within their tear samples and two sets of twins in the cohort showed exceptionally similar proteomes, supporting validity of the analysis. Conclusions: Tear sampling by Schirmer test strips and subsequent proteomic analysis by mass spectrometry in preterm infants is feasible. A larger study is required to investigate the potential use of tear proteomics in identification of ROP. Translational Relevance: Tear sampling and subsequent mass spectrometry in preterm infants is feasible. Investigation of the premature tear proteome may increase our understanding of retinal development and provide noninvasive biomarkers for identification of treatment-warranted ROP.


Subject(s)
Biomarkers , Eye Proteins , Feasibility Studies , Gestational Age , Infant, Premature , Proteomics , Retinopathy of Prematurity , Tears , Humans , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/metabolism , Proteomics/methods , Infant, Newborn , Female , Tears/chemistry , Tears/metabolism , Male , Biomarkers/metabolism , Biomarkers/analysis , Eye Proteins/metabolism , Eye Proteins/analysis , Infant , Mass Spectrometry/methods
3.
BMC Genomics ; 25(1): 415, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38671350

ABSTRACT

Oxygen-induced retinopathy (OIR) animal model is widely used for retinopathy of prematurity (ROP) researches. The purpose of this study was to identify proteins and related pathways of OIR with or without anti-vascular endothelial growth factor (VEGF) treatment, for use as biomarkers in diagnosing and treating ROP. Nine samples were subjected to proteomic analysis. Retina specimens were collected from 3 OIR mice, 3 OIR mice with anti-VEGF treatment and 3 normal mice (control group). Liquid chromatography-tandem mass spectrometry analysis was performed using the 4D label-free technique. Statistically significant differentially expressed proteins, gene ontology (GO) terms, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway representations, InterPro (IPR) and protein interactions were analyzed. In total, 4585 unique proteins were identified as differentially expressed proteins (DEPs). Enrichment analysis of the GO and KEGG indicated functional clusters related to peptide biosynthetic and metabolic process, cellular macromolecule biosynthetic process and nucleic acid binding in OIR group. For anti-VEGF treatment group, DEPs were clustered in DNA replication, PI3K/Akt signaling pathway and Jak/STAT signaling pathway. Proteomic profiling is useful for the exploration of molecular mechanisms of OIR and mechanisms of anti-VEGF treatment. These findings may be useful for identification of novel biomarkers for ROP pathogenesis and treatment.


Subject(s)
Oxygen , Proteomics , Retinopathy of Prematurity , Vascular Endothelial Growth Factor A , Animals , Oxygen/metabolism , Mice , Proteomics/methods , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Signal Transduction/drug effects , Disease Models, Animal , Tandem Mass Spectrometry , Gene Ontology , Chromatography, Liquid , Retina/metabolism , Retina/drug effects , Retina/pathology
5.
Chem Biol Drug Des ; 103(3): e14504, 2024 03.
Article in English | MEDLINE | ID: mdl-38480485

ABSTRACT

We conducted a study on the impact of intraperitoneal injections of melatonin and its three bioisosteres (compounds 1-3) on the development of oxygen-induced retinopathy in newborn rats during a 21-day experiment. It was demonstrated that melatonin and its analogues 1-3 effectively reduce the total protein concentration in the vitreous body of rat pups, decrease concentration of VEGF-A, and lower the level of oxidative stress (as indicated by normalization of antioxidant activity in the vitreous body). Melatonin and its analogues 1-3 equally normalize the level of VEGF-A. Analogues 1 and 2 even exceed melatonin in their ability to reduce protein influx into the vitreous body. However, analogue 2 had no effect on antioxidant activity, while analogues 1 and 3 caused a significant increase in this parameter, with analogue 3 even slightly exceeding melatonin. Thus, it can be concluded that analogues 1-3 are comparable to melatonin and can be utilized as potential therapeutic agents for the treatment of retinopathy of prematurity.


Subject(s)
Melatonin , Retinopathy of Prematurity , Rats , Animals , Melatonin/pharmacology , Melatonin/therapeutic use , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Disease Models, Animal
6.
Exp Eye Res ; 239: 109750, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38097102

ABSTRACT

Retinopathy of prematurity (ROP) is the leading cause of blindness in children, but there is no safe and effective treatment available. Interleukin-1 receptor type 2 (IL1R2) acts as a decoy receptor for IL-1 may affect ROP progression. This study aimed to investigate the role of IL1R2 in ROP. A microglial cell model was established under hypoxia conditions and co-cultured with choroidal endothelial cells, while an oxygen-induced retinopathy (OIR) model was also established. Microglial activation and IL1R2 levels in retinal tissues were analyzed using immunofluorescence assay. Endothelial cell migration was evaluated by Transwell assay and scratch test, angiogenesis was assessed using ELISA and tube formation assay, and proliferation was evaluated by EdU assay. The HIF1α/PFKFB3 pathway was analyzed by western blot. We observed that IL1R2 expression was predicted to be upregulated in ROP and was increased in hypoxia-treated BV2 cells. Additionally, IL1R2 levels were upregulated in the retinal tissues of OIR mice and correlated with microglial activation. In vitro experiments, we found that hypoxia promoted endothelial cell migration, angiogenesis, proliferation, and activated the HIF1α/PFKFB3 pathway, which were rescued by IL1R2 knockdown. Moreover, NHWD-870 (a HIF1α/PFKFB3 pathway inhibitor) suppressed endothelial cell migration, angiogenesis, and proliferation induced by IL1R2 overexpression. In conclusion, IL1R2 facilitates the migration, angiogenesis, and proliferation of choroidal endothelial cells by activating the HIF1α/PFKFB3 pathway to regulate ROP progression.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Animals , Humans , Mice , Angiogenesis , Disease Models, Animal , Endothelial Cells/metabolism , Hypoxia/metabolism , Mice, Inbred C57BL , Oxygen/metabolism , Phosphofructokinase-2/adverse effects , Phosphofructokinase-2/metabolism , Receptors, Interleukin-1 Type II/metabolism , Retina/metabolism , Retinal Neovascularization/metabolism , Retinopathy of Prematurity/metabolism
7.
J Neuroinflammation ; 20(1): 308, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129891

ABSTRACT

Pathological neovascularization is a pivotal biological process in wet age-related macular degeneration (AMD), retinopathy of prematurity (ROP) and proliferative diabetic retinopathy (PDR), in which macrophages (Mφs) play a key role. Tip cell specialization is critical in angiogenesis; however, its interconnection with the surrounding immune environment remains unclear. Succinate is an intermediate in the tricarboxylic acid (TCA) cycle and was significantly elevated in patients with wet AMD by metabolomics. Advanced experiments revealed that SUCNR1 expression in Mφ and M2 polarization was detected in abnormal vessels of choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR) models. Succinate-induced M2 polarization via SUCNR1, which facilitated vascular endothelial cell (EC) migration, invasion, and tubulation, thus promoting angiogenesis in pathological neovascularization. Furthermore, evidence indicated that succinate triggered the release of RBP4 from Mφs into the surroundings to regulate endothelial sprouting and pathological angiogenesis via VEGFR2, a marker of tip cell formation. In conclusion, our results suggest that succinate represents a novel class of vasculature-inducing factors that modulate Mφ polarization and the RBP4/VEGFR2 pathway to induce pathological angiogenic signaling through tip cell specialization.


Subject(s)
Choroidal Neovascularization , Retinopathy of Prematurity , Infant, Newborn , Humans , Animals , Succinic Acid/metabolism , Eye/metabolism , Choroidal Neovascularization/metabolism , Retinopathy of Prematurity/metabolism , Macrophages/metabolism , Disease Models, Animal , Retinol-Binding Proteins, Plasma/metabolism
8.
Sci Rep ; 13(1): 16175, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37759071

ABSTRACT

Deregulation of vascular endothelial growth factor (VEGF) levels leads to retinopathy of prematurity (ROP). Vitamin D (VIT-D) is known to regulate VEGF in an oxygen dependent manner. The purpose of this study was to correlate tear levels of VEGF and VIT-D with different ROP stages in preterm infants. In this prospective cross-sectional study, we enrolled 104 pre-term infants. They were grouped into: Group-1 (Classical ROP) and Group-2 (Aggressive ROP), which were further subdivided into Group-1A (progressing), Group-1B (regressing), Group-2A (pre-treatment), and Group-2B (post-treatment). Tear VEGF and VIT-D levels and their association with different ROP stages were assessed. Stage 1 and stage 2 had higher whereas stage 3 had lower VEGF levels in Group-1B compared to Group-1A. Stage 1 and stage 3 showed higher levels of VIT-D with no difference in stage 2 in Group-1B compared to Group-1A., Group-2B showed higher VEGF and lower VIT-D levels compared to Group-2A. Presence of a positive correlation at an early stage (stage 1) of ROP and a negative correlation at a more advanced stage (stage 3) of ROP with VIT-D and VEGF implies stage-specific distinct signaling crosstalk. These findings suggest that VIT-D supplementation may have the potential to modify the course and outcome of ROP.


Subject(s)
Infant, Premature , Retinopathy of Prematurity , Infant , Humans , Infant, Newborn , Vascular Endothelial Growth Factor A , Vitamin D , Prospective Studies , Retinopathy of Prematurity/metabolism , Cross-Sectional Studies , Gestational Age
9.
Sci Rep ; 13(1): 13319, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37587267

ABSTRACT

Retinopathy of Prematurity (ROP) is a multifactorial disease characterized by abnormal retinal vascular growth in premature infants, which is one of the leading causes of childhood blindness. Lactic acid metabolism may play an imperative role in the development of ROP, but there are still few relevant studies. Our team use a dataset GSE158799 contained 284 genes in 3 P17_OIR mice and 3 P30_OIR mice to identify 41 potentially differentially expressed lactate metabolism-related genes (LMRGs) related to ROP. Then through bioinformatics analysis, we strive to reveal the interaction, the enriched pathways and the immune cell infiltration among these LMRGs, and predict their functions and internal mechanisms. These DEGs may regulate lactate metabolism, leading to the changes of metabolism and immunity, thereby inducing the development of ROP. Our results will expand our understanding of the intrinsic mechanism of ROP and may be helpful for the directions for treatment of ROP in the future.


Subject(s)
Gene Expression Regulation , Retinopathy of Prematurity , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism , Oxygen/toxicity , Animals , Mice , Mice, Inbred C57BL , Lactic Acid/metabolism , Signal Transduction
10.
Invest Ophthalmol Vis Sci ; 64(11): 17, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37566408

ABSTRACT

Purpose: Retinal ischemia is a common cause of a variety of eye diseases, such as retinopathy of prematurity, diabetic retinopathy, and vein occlusion. Protein kinase RNA-activated-like endoplasmic reticulum (ER) kinase (PERK), one of the main ER stress sensor proteins, has been involved in many diseases. In this study, we investigated the role of PERK in ischemia-induced retinopathy using a mouse model of oxygen-induced retinopathy (OIR). Methods: OIR was induced by subjecting neonatal pups to 70% oxygen at postnatal day 7 (P7) followed by returning to room air at P12. GSK2606414, a selective PERK inhibitor, was orally administrated to pups right after they were returned to room air once daily until 1 day before sample collection. Western blot, immunostaining, and quantitative PCR were used to assess PERK phosphorylation, retinal changes, and signaling pathways in relation to PERK inhibition. Results: PERK phosphorylation was prominently increased in OIR retinas, which was inhibited by GSK2606414. Concomitantly, PERK inhibition significantly reduced retinal neovascularization (NV) and retinal ganglion cell (RGC) loss, restored astrocyte network, and promoted revascularization. Furthermore, PERK inhibition downregulated the recruitment/proliferation of mononuclear phagocytes but did not affect OIR-upregulated canonical angiogenic pathways. Conclusions: Our results demonstrate that PERK is involved in ischemia-induced retinopathy and its inhibition using GSK2606414 could offer an effective therapeutic intervention aimed at alleviating retinal NV while preventing neuron loss during retinal ischemia.


Subject(s)
Retinal Diseases , Retinal Neovascularization , Retinopathy of Prematurity , eIF-2 Kinase , Animals , Mice , Animals, Newborn , Disease Models, Animal , Ischemia/metabolism , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Oxygen/metabolism , Retina , Retinal Diseases/etiology , Retinal Diseases/prevention & control , Retinal Ganglion Cells/metabolism , Retinal Neovascularization/prevention & control , Retinal Neovascularization/metabolism , Retinopathy of Prematurity/metabolism , eIF-2 Kinase/metabolism
11.
Exp Eye Res ; 233: 109547, 2023 08.
Article in English | MEDLINE | ID: mdl-37348672

ABSTRACT

Despite decades of researches, the underlying mechanism of retinopathy of prematurity (ROP) remains unclear. The role of Sirt2, which is involved in both angiogenesis and inflammation, both pivotal in ROP, was investigated in an animal model of ROP known as oxygen-induced retinopathy (OIR). Our study found that Sirt2 was overexpressed and colocalized with microglia in OIR. Furthermore, it demonstrated that the level of Sirt2 was upregulated in hypoxia microglia BV-2 in vitro. Subsequently, our results elucidated that administration of the Sirt2 antagonist AGK2 attenuated the avascular and neovascular area and downregulated the expression of IGF-1. The phosphorylation of Akt and the expression of IGF-1 were upregulated in hypoxia BV-2 and conditional media collected from BV-2 under hypoxia promoted the migration and tube formation of retinal capillary endothelial cells, which were suppressed with AGK2. Notably, our findings are the first to demonstrate the deleterious role of Sirt2 in ROP, as Sirt2 inhibition led to the downregulation of Akt/IGF-1 and ameliorated vasculopathy, ultimately improving visual function. These results suggest that Sirt2 may be a promising therapeutic target for ROP.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Animals , Humans , Infant, Newborn , Mice , Retinopathy of Prematurity/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Insulin-Like Growth Factor I/adverse effects , Insulin-Like Growth Factor I/metabolism , Endothelial Cells/metabolism , Sirtuin 2/genetics , Retinal Neovascularization/metabolism , Oxygen/toxicity , Hypoxia , Disease Models, Animal , Mice, Inbred C57BL , Animals, Newborn
12.
Indian J Pediatr ; 90(11): 1089-1095, 2023 11.
Article in English | MEDLINE | ID: mdl-37227582

ABSTRACT

OBJECTIVE: To determine whether red blood cell glucose-6-phosphate dehydrogenase (G6PD) activity is associated with retinopathy of prematurity (ROP). METHODS: This case-control study was conducted in a Level-3 neonatal unit. Subjects were inborn boys with birth weight <2000 g. "Cases" were consecutive subjects with ROP of any severity. "Controls" were consecutive unrelated subjects without ROP. Recipients of blood or exchange transfusions were excluded. Sixty cases (out of 98 screened) and 60 controls (out of 93 screened) were enrolled. G6PD activity (quantitative assay) as the candidate risk factor was evaluated. RESULTS: Sixty cases with 60 controls [mean (SD) gestation 28.80 (2.2) and 30.60 (2.2) wk respectively] were compared. "Cases" had a higher median (1st, 3rd quartile) G6PD activity compared to "controls" [7.39 (4.7, 11.5) vs. 6.28 (4.2, 8.8) U/g Hb, p = 0.084]. G6PD activity was highest among ROP requiring treatment [8.68 (4.7, 12.3)] followed by ROP not requiring treatment [6.91 (4.4, 11.0)], followed by controls (plinear trend = 0.06). Gestation, birth weight, duration of oxygen, breastmilk feeding, and clinical sepsis were other variables associated with ROP on univariable analysis. On multivariable logistic regression, G6PD activity [Adjusted OR 1.14 (1.03, 1.25), p = 0.01] and gestation [Adjusted OR 0.74 (0.56, 0.97), p = 0.03] independently predicted ROP. C-statistic of the model was 0.76 (95% CI 0.67, 0.85). CONCLUSIONS: Higher G6PD activity was independently associated with ROP after adjusting for confounders. Each 1 U/g Hb increase in G6PD increased the odds of ROP by 14%. Severer forms of ROP were associated with higher levels of G6PD activity.


Subject(s)
Glucosephosphate Dehydrogenase , Retinopathy of Prematurity , Humans , Infant, Newborn , Male , Birth Weight , Case-Control Studies , Gestational Age , Infant, Premature , Retinopathy of Prematurity/epidemiology , Retinopathy of Prematurity/etiology , Retinopathy of Prematurity/metabolism , Retrospective Studies , Risk Factors
13.
Exp Eye Res ; 232: 109518, 2023 07.
Article in English | MEDLINE | ID: mdl-37257714

ABSTRACT

The aim of the study is to reveal the expression profiling and clinical significance of peripheral blood mononuclear cell (PBMC) tRNA-derived small RNAs (tsRNAs) and microRNAs (miRNAs) of premature infants with treatment-requiring retinopathy of prematurity (ROP). Significantly altered tsRNAs and miRNAs were screened using small RNA sequencing. RT-qPCR was used to verify the altered RNAs identified by small RNA transcriptomics. The target genes, their enriched functions, and possibly involved signaling pathways were identified by bioinformatics analyses. According to the small RNA sequencing, 125 tsRNAs and 205 miRNAs were significantly altered in PBMCs obtained from infants with treatment-requiring ROP compared with the premature controls without retinopathy. We preliminarily validated the significant alterations of 6 tsRNAs and 9 miRNAs. The target genes for those tsRNAs were enriched for cellular macromolecule metabolic process, intracellular anatomical structure, transcription regulatory region nucleic acid binding, and Th17 cell differentiation; those of the altered miRNAs were enriched for the developmental process, cell junction, DNA-binding transcription activator activity, and FoxO signaling pathway. By verification with the extended sample size, we identified tsRNAs and miRNAs that could be potential biomarkers with clinical values. The study recognized the alterations and clinical significance of changed tsRNA/miRNA profiles in PBMCs from premature infants with ROP. These significantly altered tsRNAs and miRNAs might be useful as potential diagnostic biomarkers and molecular targets for treatment-requiring ROP.


Subject(s)
MicroRNAs , Retinopathy of Prematurity , Infant, Newborn , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Leukocytes, Mononuclear/metabolism , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism , Clinical Relevance , Biomarkers/metabolism
14.
Graefes Arch Clin Exp Ophthalmol ; 261(9): 2477-2488, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37022494

ABSTRACT

PURPOSE: To determine whether 14 inflammation-, angiogenesis-, and adhesion-related proteins in cord blood (CB), alone or in combination with conventional perinatal factors, could predict retinopathy of prematurity (ROP) in preterm infants. METHODS: Data from 111 preterm infants (born at ≤ 32.0 weeks) were retrospectively reviewed. The levels of endoglin, E-selectin, HSP70, IGFBP-3/4, LBP, lipocaline-2, M-CSFR, MIP-1α, pentraxin 3, P-selectin, TGFBI, TGF-ß1, and TNFR2 were assessed in stored CB samples collected at birth using ELISA kits. The primary endpoints included severe ROP (≥ stage 3) and type 1 ROP requiring treatment. RESULTS: ROP was diagnosed in 29 infants (26.1%), among whom 14 (12.6%) had severe ROP and seven (6.3%) had type 1 ROP. Multivariate logistic regression showed that decreased CB TGFBI levels were significantly associated with severe ROP and type 1 ROP after adjusting for gestational age at birth. Stepwise regression analysis allowed to design prediction models with good accuracy, which comprised low CB TGFBI levels and low birth weight (BW) as predictors for severe ROP (area under the curve [AUC] = 0.888), and low CB endoglin levels and low BW as predictors for type 1 ROP (AUC = 0.950). None of the other CB proteins evaluated were found to be associated with severe ROP or type 1 ROP. CONCLUSIONS: Low CB TGFBI levels are associated with severe ROP and type 1 ROP, independently of gestational age. Moreover, combined predictive models based on CB TGFBI and endoglin levels, along with BW data, may act as good indicators at birth for the neonatal risk of ROP progression.


Subject(s)
Infant, Premature , Retinopathy of Prematurity , Infant , Pregnancy , Female , Infant, Newborn , Humans , Retrospective Studies , Transforming Growth Factor beta , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/metabolism , Fetal Blood/metabolism , Endoglin , Risk Factors , Gestational Age , Biomarkers , Transforming Growth Factors , Birth Weight
15.
PLoS One ; 18(4): e0284764, 2023.
Article in English | MEDLINE | ID: mdl-37098032

ABSTRACT

BACKGROUND: Anemia and retinopathy of prematurity (ROP) are common comorbidities experienced by preterm infants, yet the role of anemia on the pathogenesis of ROP remains unclear. Reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) is a sensitive technique for estimating the gene expression changes at the transcript level but requires identification of stably expressed reference genes for accurate data interpretation. This is particularly important for oxygen induced retinopathy studies given that some commonly used reference genes are sensitive to oxygen. This study aimed to identify stably expressed reference genes among eight commonly used reference genes in the neonatal rat pups' retina upon exposure to cyclic hyperoxia-hypoxia, anemia, and erythropoietin administration at two age groups (P14.5 and P20) using Bestkeeper, geNorm, and Normfinder, three publicly available, free algorithms, and comparing their results to the in-silico prediction program, RefFinder. RESULTS: The most stable reference gene across both developmental stages was Rpp30, as predicted by Genorm, Bestkeeper, and Normfinder. RefFinder predicted Tbp to be the most stable across both developmental stages. At P14.5, stability varied by prediction program; at P20, RPP30 and MAPK1 were the most stable reference genes. Gapdh, 18S, Rplp0, and HPRT were predicted as the least stable reference genes by at least one of the prediction algorithms. CONCLUSION: Expression of Rpp30 is the least affected by experimental conditions of oxygen induced retinopathy, phlebotomy induced anemia and erythropoietin administration at both timepoints of P14.5 and P20.


Subject(s)
Anemia , Erythropoietin , Retinopathy of Prematurity , Infant, Newborn , Humans , Rats , Animals , Oxygen/metabolism , Infant, Premature , Retina/metabolism , Retinopathy of Prematurity/chemically induced , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism , Erythropoietin/genetics , Erythropoietin/metabolism , Anemia/chemically induced , Anemia/genetics , Anemia/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reference Standards , Gene Expression Profiling/methods , Real-Time Polymerase Chain Reaction/methods
16.
J Neuroinflammation ; 20(1): 14, 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36691048

ABSTRACT

BACKGROUND: We have investigated the efficacy of a new strategy to limit pathological retinal neovascularization (RNV) during ischemic retinopathy by targeting the cholesterol metabolizing enzyme acyl-coenzyme A: cholesterol transferase 1 (ACAT1). Dyslipidemia and cholesterol accumulation have been strongly implicated in promoting subretinal NV. However, little is known about the role of cholesterol metabolism in RNV. Here, we tested the effects of inhibiting ACAT1 on pathological RNV in the mouse model of oxygen-induced retinopathy (OIR). METHODS: In vivo studies used knockout mice that lack the receptor for LDL cholesterol (LDLR-/-) and wild-type mice. The wild-type mice were treated with a specific inhibitor of ACAT1, K604 (10 mg/kg, i.p) or vehicle (PBS) during OIR. In vitro studies used human microglia exposed to oxygen-glucose deprivation (OGD) and treated with the ACAT1 inhibitor (1 µM) or PBS. RESULTS: Analysis of OIR retinas showed that increased expression of inflammatory mediators and pathological RNV were associated with significant increases in expression of the LDLR, increased accumulation of neutral lipids, and formation of toxic levels of cholesterol ester (CE). Deletion of the LDLR completely blocked OIR-induced RNV and significantly reduced the AVA. The OIR-induced increase in CE formation was accompanied by significant increases in expression of ACAT1, VEGF and inflammatory factors (TREM1 and MCSF) (p < 0.05). ACAT1 was co-localized with TREM1, MCSF, and macrophage/microglia makers (F4/80 and Iba1) in areas of RNV. Treatment with K604 prevented retinal accumulation of neutral lipids and CE formation, inhibited RNV, and decreased the AVA as compared to controls (p < 0.05). The treatment also blocked upregulation of LDLR, ACAT1, TREM1, MCSF, and inflammatory cytokines but did not alter VEGF expression. K604 treatment of microglia cells also blocked the effects of OGD in increasing expression of ACAT1, TREM1, and MCSF without altering VEGF expression. CONCLUSIONS: OIR-induced RNV is closely associated with increases in lipid accumulation and CE formation along with increased expression of LDLR, ACAT1, TREM1, and MCSF. Inhibiting ACAT1 blocked these effects and limited RNV independently of alterations in VEGF expression. This pathway offers a novel strategy to limit vascular injury during ischemic retinopathy.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Infant, Newborn , Animals , Humans , Mice , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/prevention & control , Retinopathy of Prematurity/metabolism , Triggering Receptor Expressed on Myeloid Cells-1 , Vascular Endothelial Growth Factor A/metabolism , Oxygen/metabolism , Cholesterol , Transferases , Coenzyme A/adverse effects , Lipids/adverse effects , Mice, Inbred C57BL , Disease Models, Animal , Acetyl-CoA C-Acetyltransferase
17.
Front Immunol ; 13: 953812, 2022.
Article in English | MEDLINE | ID: mdl-36081509

ABSTRACT

Purpose: This study aims to reveal the altered expression profiles of circular RNAs (circRNAs) in the peripheral blood mononuclear cells (PBMCs) of patients with retinopathy of prematurity (ROP), and to identify potential biomarkers for ROP diagnosis. Methods: Differentially expressed circRNAs in PBMCs of five infants with ROP and five controls were identified using microarray analysis. Twelve altered circRNAs were validated using reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR). Bioinformatic analyses were conducted to predict the circRNA/miRNA interactions, competing endogenous RNA (ceRNA) network, related biological functions, and signaling pathways. Four selected circRNAs in PBMCs were verified using RT-qPCR in another cohort, including 24 infants with ROP and 23 premature controls, and receiver operating characteristic (ROC) curves were used to estimate their potential as diagnostic biomarkers of ROP. Results: A total of 54 and 143 circRNAs were significantly up- and down-regulated, respectively, in the PBMCs of patients with ROP compared with controls. Twelve of the significantly altered circRNAs were preliminarily validated by RT-qPCR, which confirmed the reliability of the microarray analysis. The circRNA/miRNA interactions and ceRNA network were displayed according to the altered circRNAs. Three circRNAs (hsa_circRNA_061346, hsa_circRNA_092369, and hsa_circRNA_103554) were identified as potential diagnostic biomarkers for ROP with certain clinical values. Conclusions: CircRNAs were significantly altered in PBMCs of treatment-requiring ROP patients. CircRNAs may be used as potential biomarkers and possible therapeutic targets for ROP.


Subject(s)
Leukocytes, Mononuclear , MicroRNAs , RNA, Circular , Retinopathy of Prematurity , Biomarkers , Humans , Infant, Newborn , Leukocytes, Mononuclear/metabolism , MicroRNAs/genetics , RNA, Circular/genetics , Reproducibility of Results , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism
18.
Cell Death Dis ; 13(8): 745, 2022 08 29.
Article in English | MEDLINE | ID: mdl-36038541

ABSTRACT

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.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Animals , Arginase/genetics , Arginase/metabolism , Disease Models, Animal , Humans , Infant, Newborn , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Oxygen , Polyethylene Glycols/therapeutic use , Retinal Neovascularization/pathology , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology
19.
BMC Ophthalmol ; 22(1): 342, 2022 Aug 11.
Article in English | MEDLINE | ID: mdl-35953806

ABSTRACT

BACKGROUND: Although several clinical studies have analysed the relationship between the levels of vascular endothelial growth factor (VEGF) and apelin-13 in venous blood and retinopathy of prematurity (ROP), no definitive conclusions have been reached. This study aimed to investigate the relationship between apelin-13 levels and VEGF levels and ROP. METHODS: Differences in plasma apelin-13 and VEGF levels were analysed in two groups of infants born with birth weight < 1500 g and gestational age < 32 weeks at Peking University People' s Hospital. One group comprised infants diagnosed with ROP and the other group was a control group comprising infants without ROP. RESULTS: Apelin-13 levels were significantly lower in the ROP group than in the control group, while VEGF levels showed the opposite result (both P < 0.001). Infants with severe ROP had lower apelin-13 levels and higher VEGF levels than with mild ROP (both P < 0.05).The receiver operating characteristic curve for apelin-13 level as the indicator of ROP showed that a cut-off value of 119.6 pg/mL yielded a sensitivity of 84.8% and a specificity of 63.6%, while for VEGF level, the cut-off value of 84.3 pg/mL exhibited a sensitivity of 84.8% and a specificity of 66.7%. CONCLUSIONS: Plasma apelin-13 and VEGF levels at 4-6 weeks of age may play a role in assisting the diagnosis of ROP.


Subject(s)
Retinopathy of Prematurity , Apelin , Case-Control Studies , Gestational Age , Humans , Infant , Infant, Newborn , Infant, Very Low Birth Weight , Retinopathy of Prematurity/diagnosis , Retinopathy of Prematurity/metabolism , Vascular Endothelial Growth Factor A/metabolism
20.
Int J Mol Sci ; 23(13)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35806359

ABSTRACT

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.


Subject(s)
Retinal Neovascularization , Retinopathy of Prematurity , Animals , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Infant, Newborn , Neovascularization, Pathologic/metabolism , RNA-Seq , Rats , Rats, Sprague-Dawley , Retinal Neovascularization/metabolism , Retinal Vessels/metabolism , Retinopathy of Prematurity/genetics , Retinopathy of Prematurity/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL