Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 409
Filter
1.
Biomed Pharmacother ; 178: 117222, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39088968

ABSTRACT

BACKGROUND: Ocular neovascular diseases, which contribute significantly to vision loss, lack effective preventive treatments. Recent studies have highlighted the significant involvement of immune cells in neovascular retinopathy. Myeloid-derived suppressor cells (MDSCs) promote the development of neovascularization, but it is unknown whether they participate in pathological neovascularization and whether they are expected to be a therapeutic target. METHOD: We investigated the role of MDSCs in promoting pathological angiogenesis using an oxygen-induced retinopathy (OIR) model, employing flow cytometry, immunofluorescence, and smart-seq analysis. Then, we evaluated the proportion of MDSCs in patient blood samples using flow cytometry. Additionally, we assessed the effect of MDSC depletion using an anti-Gr-1 monoclonal antibody on retinal vasculopathy and alterations in retinal microglia. RESULTS: In the OIR model, an elevated ratio of MDSCs was observed in both blood and retinal tissue during phase II (Neovascularization). The depletion of MDSCs resulted in reduced retinal neovascularization and vaso-obliteration, along with a decrease in microglia within the neovascularization area. Furthermore, analysis of gene transcripts associated with MDSCs indicated activation of vascular endothelial growth factor (VEGF) regulation and inflammation. Importantly, infants with ROP exhibited a higher proportion of MDSCs in their blood samples. CONCLUSION: Our results suggested that excessive MDSCs represent an unrecognized feature of ocular neovascular diseases and be responsible for the retinal vascular inflammation and angiogenesis, providing opportunities for new therapeutic approaches to ocular neovascular disease.


Subject(s)
Myeloid-Derived Suppressor Cells , Retinal Neovascularization , Humans , Myeloid-Derived Suppressor Cells/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/metabolism , Animals , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor A/metabolism , Mice , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Microglia/pathology , Microglia/metabolism , Infant , Oxygen/blood
2.
Cells ; 13(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39195259

ABSTRACT

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


Subject(s)
Angiogenesis Inhibitors , Anti-Inflammatory Agents , Disease Models, Animal , Oxygen , Retinopathy of Prematurity , Animals , Mice , Humans , Oxygen/metabolism , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Anti-Inflammatory Agents/pharmacology , Mice, Inbred C57BL , Retinal Neovascularization/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/drug therapy , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/antagonists & inhibitors , Cytokines/metabolism , Hyperoxia/complications , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 2/antagonists & inhibitors , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Small Molecule Libraries/pharmacology
3.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39126030

ABSTRACT

Retinopathy of prematurity (ROP) is a vascular disorder affecting the retinas of preterm infants. This condition arises when preterm infants in incubators are exposed to high oxygen levels, leading to oxidative stress, inflammatory responses, and a downregulation of vascular endothelial growth factors, which causes the loss of retinal microvascular capillaries. Upon returning to room air, the upregulation of vascular growth factors results in abnormal vascular growth of retinal endothelial cells. Without appropriate intervention, ROP can progress to blindness. The prevalence of ROP has risen, making it a significant cause of childhood blindness. Current treatments, such as laser therapy and various pharmacologic approaches, are limited by their potential for severe adverse effects. Therefore, a deeper understanding of ROP's pathophysiology and the development of innovative treatments are imperative. Natural products from plants, fungi, bacteria, and marine organisms have shown promise in treating various diseases and have gained attention in ROP research due to their minimal side effects and wide-ranging beneficial properties. This review discusses the roles and mechanisms of natural products that hold potential as therapeutic agents in ROP management.


Subject(s)
Biological Products , Retinopathy of Prematurity , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/therapy , Retinopathy of Prematurity/metabolism , Humans , Biological Products/therapeutic use , Biological Products/pharmacology , Infant, Newborn , Animals , Infant, Premature
4.
Invest Ophthalmol Vis Sci ; 65(10): 34, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39186263

ABSTRACT

Purpose: In response to hypoxia, sympathetic fibers to the retina activate ß-adrenoceptors (ß-ARs) that play an important role in the regulation of vascular and neuronal functions. We investigated the role of ß3-AR using the mouse model of oxygen-induced retinopathy (OIR). Methods: Mouse pups were exposed to 75% oxygen at postnatal day 7 (PD7) followed by a return to room air at PD12. The ß3-AR preferential agonist BRL37344 was subcutaneously administered once daily at different times after the return to room air. At PD17, the OIR mice underwent flash and pattern electroretinogram. After sacrifice, retinal wholemounts were used for vessel staining or immunohistochemistry for astrocytes, Müller cells, or retinal ganglion cells (RGCs). In retinal homogenates, the levels of markers associated with neovascularization (NV), the blood-retinal barrier (BRB), or astrocytes were determined by western blot, and quantitative reverse-transcription polymerase chain reaction was used to assess ß3-AR messenger. Administration of the ß3-AR antagonist SR59230A was performed to verify BRL37344 selectivity. Results: ß3-AR expression is upregulated in response to hypoxia, but its increase is prevented by BRL37344, which counteracts NV by inhibiting the pro-angiogenic pathway, activating the anti-angiogenic pathway, recovering BRB-associated markers, triggering nitric oxide production, and favoring revascularization of the central retina through recovered density of astrocytes that ultimately counteracts NV in the midperiphery. Vasculature rescue prevents dysfunctional retinal activity and counteracts OIR-associated retinal ganglion cell loss. Conclusions: ß3-AR has emerged as a crucial intermediary in hypoxia-dependent NV, suggesting a role of ß3-AR agonists in the treatment of proliferative retinopathies.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Disease Models, Animal , Electroretinography , Mice, Inbred C57BL , Oxygen , Receptors, Adrenergic, beta-3 , Retinal Neovascularization , Animals , Mice , Retinal Neovascularization/metabolism , Retinal Neovascularization/prevention & control , Retinal Neovascularization/pathology , Oxygen/toxicity , Adrenergic beta-3 Receptor Agonists/pharmacology , Receptors, Adrenergic, beta-3/metabolism , Animals, Newborn , Retinal Ganglion Cells/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/metabolism , Ethanolamines/pharmacology , Retinal Vessels/drug effects , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/drug therapy , Astrocytes/metabolism , Astrocytes/drug effects , Immunohistochemistry , Angiogenesis
5.
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)
Oxygen , Retinal Neovascularization , Retinopathy of Prematurity , Animals , Humans , Mice , Animals, Newborn , Disease Models, Animal , Hyperoxia/complications , Intravitreal Injections , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Mice, Inbred C57BL , Oxygen/toxicity , Retinal Neovascularization/metabolism , Retinal Neovascularization/prevention & control , Retinal Neovascularization/pathology , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
6.
Angiogenesis ; 27(3): 423-440, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38709389

ABSTRACT

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.


Subject(s)
Oxygen , Retinopathy of Prematurity , Animals , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Mice , Antibodies, Monoclonal, Humanized/pharmacology , Mice, Inbred C57BL , Animals, Newborn , Disease Models, Animal , Humans , Hyperoxia/pathology , Hyperoxia/complications , Retina/pathology , Retina/metabolism , Retina/drug effects , CARD Signaling Adaptor Proteins/metabolism , Mice, Transgenic , Retinal Neovascularization/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/drug therapy , Microglia/pathology , Microglia/metabolism , Microglia/drug effects
7.
Immunology ; 173(1): 141-151, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38804253

ABSTRACT

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.


Subject(s)
Flavonoids , Hexokinase , Microglia , Oxygen , Retinal Neovascularization , Retinopathy of Prematurity , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Humans , Mice , Cell Movement/drug effects , Disease Models, Animal , Flavonoids/pharmacology , Flavonoids/therapeutic use , Hexokinase/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Oxygen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Retinal Neovascularization/drug therapy , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
8.
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
10.
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
11.
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
12.
Angiogenesis ; 27(3): 379-395, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38483712

ABSTRACT

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.


Subject(s)
ADAM17 Protein , Proto-Oncogene Proteins c-fos , Retinal Neovascularization , Animals , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Retinal Neovascularization/genetics , ADAM17 Protein/metabolism , ADAM17 Protein/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , Mice , Humans , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology , Retinopathy of Prematurity/genetics , Mice, Inbred C57BL , Transcription, Genetic , Gene Expression Regulation , Retinal Vessels/metabolism , Retinal Vessels/pathology , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Disease Models, Animal , Angiogenesis
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL