Abstract

BackgroundAbnormal neovascularization is the most common cause of blindness, and hypoxia alters tissue metabolism, function, and morphology. HIF-1α, the transcriptional activator of VEGF, has intricate mechanisms of nuclear translocation and activation, but its signal termination mechanisms remain unclear.MethodsWe investigated the role of polypyrimidine tract-binding protein-associated splicing factor (PSF) in cellular energy production, migration, and proliferation by targeting HIF-1α in vivo and in vitro PSF plasmids were transfected with liposome 2000 transfection reagent. Young C57/BL6J mice were kept in a hyperoxia environment, followed by indoor air, resulting in oxygen-induced retinopathy. Oxygen-induced retinopathy (OIR) animals were randomly divided into three groups: OIR group, OIR + vector group (OIR cubs treated with rAAV vector) and OIR + PSF group (OIR cubs treated with rAAV-PSF). Age-matched C57/BL6J mice were used as controls and exposed to constant normoxic conditions. The animals were executed and their pupils were subjected to subsequent experiments. The metabolic spectrum was analyzed by Seahorse XFe96 flux analyzer, and OCR and extracellular acidification rate were quantified at the same time.ResultsPSF ameliorated retinal neovascularization and corrected abnormal VEGF expression in mice with oxygen-induced retinopathy and reduced intra-retinal neovascularization in Vldlr − / − mice. PSF reprogrammed mitochondrial bioenergetics and inhibited the transition of endothelial cells after hypoxia, suggesting its involvement in pathological angiogenesis.Ectopic PSF expression inhibited hypoxia-induced HIF-1α activation in the nucleus by recruiting Hakai to the PSF/HIF-1α complex, causing HIF-1α inhibition. PSF knockdown increased hypoxia-stimulated HIF-1α reactions. These hypoxia-dependent processes may play a vital role in cell metabolism, migration, and proliferation. Thus, PSF is a potential treatment target in neovascularization-associated ophthalmopathy.ConclusionThis is the first study showing that PSF inhibits HIF-1α via recruitment of Hakai, modulates mitochondrial oxidation and glycolysis, and downregulates VEGF expression under hypoxia. We propose a new HIF-1 α/Hakai regulatory mechanism that may play a vital role in the pathogenesis of neovascularization in ophthalmopathy.PSF-Hakai–HIF-1α signaling pathway under hypoxia condition. Schematic diagram showing that the PSF-Hakai–HIF-1α signaling pathway. Under hypoxia condition, PSF-Hakai complex regulate HIF-1α signaling, thus inhibiting downstream target gene VEGF, cell metabolism and angiogenesis eventually.Bpb6gLDdM8K1hTjkEP-G9vVideo : Detailed information of Materials and Methods.

Highlights

  • Abnormal neovascularization is the most common cause of blindness, and hypoxia alters tissue metabolism, function, and morphology

  • Our results emphasize the relationship between the protein-associated splicing factor (PSF)-Hakai–Hypoxia-inducible factor-1 alpha (HIF-1α) signaling pathways, mitochondrial function, human retinal microcapillary endothelial cells (HRMECs) proliferation and migration related to pathologic neovascularization

  • PSF improved retinal neovascularization (RNV) and rectified abnormal vascular endothelial growth factor (VEGF) production in an oxygen‐induced retinopathy (OIR) model To examine the importance of PSF in the RNV process, we used the well-established Oxygen-induced retinopathy (OIR) mouse model to imitate pathologic development of RNV and treated them with recombinant adeno-associated virus or rAAVPSF particles intravitreally on postnatal (P) day 12 and P15

Read more

Summary

Introduction

Abnormal neovascularization is the most common cause of blindness, and hypoxia alters tissue metabolism, function, and morphology. Hypoxia can lead to a series of pathological processes and cause anomalous changes in tissues and organs with respect to metabolism, function, and morphological structure [1,2,3]; As an upstream mediator of vascular endothelial growth factor (VEGF), hypoxia-inducible factor-1 alpha (HIF-1α) is a significant transcription factor for cellular response in hypoxia, and promotes angiogenesis and boosts metabolism, provoking abnormal vascularization [4,5,6]. Hakai is involved in the control of cell proliferation [11] and promotes the expression of cancer-related genes [14]. Hakai can affect cell proliferation and oncogene phenotype by improving the ability of PSF to bind RNA, thereby promoting cancer-related genes expression [16]. We hypothesized that the PSF-Hakai complex may have beneficial effects on neovascularization via HIF-1α inhibition

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call