Hypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axis

Abstract Background Pathological neovascularization plays a pivotal role in the onset and progression of tumors and neovascular eye diseases. Despite notable advancements in the development of anti-angiogenic medications that target vascular endothelial growth factor (VEGF) and its receptors (VEGFRs...

Full description

Bibliographic Details
Main Authors: Jun-Song Ren, Wen Bai, Jing-Juan Ding, Hui-Min Ge, Su-Yu Wang, Xi Chen, Qin Jiang
Format: Article
Language:English
Published: BMC 2023-09-01
Series:Journal of Translational Medicine
Subjects:
Online Access:https://doi.org/10.1186/s12967-023-04503-x
_version_ 1797556846667497472
author Jun-Song Ren
Wen Bai
Jing-Juan Ding
Hui-Min Ge
Su-Yu Wang
Xi Chen
Qin Jiang
author_facet Jun-Song Ren
Wen Bai
Jing-Juan Ding
Hui-Min Ge
Su-Yu Wang
Xi Chen
Qin Jiang
author_sort Jun-Song Ren
collection DOAJ
description Abstract Background Pathological neovascularization plays a pivotal role in the onset and progression of tumors and neovascular eye diseases. Despite notable advancements in the development of anti-angiogenic medications that target vascular endothelial growth factor (VEGF) and its receptors (VEGFRs), the occurrence of adverse reactions and drug resistance has somewhat impeded the widespread application of these drugs. Therefore, additional investigations are warranted to explore alternative therapeutic targets. In recent years, owing to the swift advancement of high-throughput sequencing technology, pan-cancer analysis and single-cell sequencing analysis have emerged as pivotal methodologies and focal areas within the domain of omics research, which is of great significance for us to find potential targets related to the regulation of pathological neovascularization. Methods Pan-cancer analysis and scRNA-seq data analysis were employed to forecast the association between Actin filament-associated protein 1 like 1 (AFAP1L1) and the development of tumors and endothelial cells. Tumor xenograft model and ocular pathological neovascularization model were constructed as well as Isolectin B4 (IsoB4) staining and immunofluorescence staining were used to assess the effects of AFAP1L1 on the progression of neoplasms and neovascular eye diseases in vivo. Transwell assay, wound scratch assay, tube forming assay, three-dimensional germination assay, and rhodamine-phalloidin staining were used to evaluate the impact of AFAP1L1 on human umbilical vein endothelial cells (HUVECs) function in vitro; Dual luciferase reporting, qRT-PCR and western blot were used to investigate the upstream and downstream mechanisms of pathological neovascularization mediated by AFAP1L1. Results Our investigation revealed that AFAP1L1 plays a crucial role in promoting the development of various tumors and demonstrates a strong correlation with endothelial cells. Targeted suppression of AFAP1L1 specifically in endothelial cells in vivo proves effective in inhibiting tumor formation and ocular pathological neovascularization. Mechanistically, AFAP1L1 functions as a hypoxia-related regulatory protein that can be activated by HIF-1α. In vitro experiments demonstrated that reducing AFAP1L1 levels can reverse hypoxia-induced excessive angiogenic capacity in HUVECs. The principal mechanism of angiogenesis inhibition entails the regulation of tip cell behavior through the YAP-DLL4-NOTCH axis. Conclusion In conclusion, AFAP1L1, a newly identified hypoxia-related regulatory protein, can be activated by HIF-1α. Inhibiting AFAP1L1 results in the inhibition of angiogenesis by suppressing the germination of endothelial tip cells through the YAP-DLL4-NOTCH axis. This presents a promising therapeutic target to halt the progression of tumors and neovascular eye disease.
first_indexed 2024-03-10T17:07:49Z
format Article
id doaj.art-5a5e734bdf534ce2a2adeff6c567d7d3
institution Directory Open Access Journal
issn 1479-5876
language English
last_indexed 2024-03-10T17:07:49Z
publishDate 2023-09-01
publisher BMC
record_format Article
series Journal of Translational Medicine
spelling doaj.art-5a5e734bdf534ce2a2adeff6c567d7d32023-11-20T10:44:39ZengBMCJournal of Translational Medicine1479-58762023-09-0121112510.1186/s12967-023-04503-xHypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axisJun-Song Ren0Wen Bai1Jing-Juan Ding2Hui-Min Ge3Su-Yu Wang4Xi Chen5Qin Jiang6Department of Ophthalmology, The Affiliated Eye Hospital, Nanjing Medical UniversityDepartment of Ophthalmology, The Affiliated Eye Hospital, Nanjing Medical UniversityDepartment of Ophthalmology, Xuzhou Central HospitalDepartment of Ophthalmology, The Affiliated Eye Hospital, Nanjing Medical UniversityDepartment of Ophthalmology, The Affiliated Eye Hospital, Nanjing Medical UniversityDepartment of Ophthalmology, The Affiliated Eye Hospital, Nanjing Medical UniversityDepartment of Ophthalmology, The Affiliated Eye Hospital, Nanjing Medical UniversityAbstract Background Pathological neovascularization plays a pivotal role in the onset and progression of tumors and neovascular eye diseases. Despite notable advancements in the development of anti-angiogenic medications that target vascular endothelial growth factor (VEGF) and its receptors (VEGFRs), the occurrence of adverse reactions and drug resistance has somewhat impeded the widespread application of these drugs. Therefore, additional investigations are warranted to explore alternative therapeutic targets. In recent years, owing to the swift advancement of high-throughput sequencing technology, pan-cancer analysis and single-cell sequencing analysis have emerged as pivotal methodologies and focal areas within the domain of omics research, which is of great significance for us to find potential targets related to the regulation of pathological neovascularization. Methods Pan-cancer analysis and scRNA-seq data analysis were employed to forecast the association between Actin filament-associated protein 1 like 1 (AFAP1L1) and the development of tumors and endothelial cells. Tumor xenograft model and ocular pathological neovascularization model were constructed as well as Isolectin B4 (IsoB4) staining and immunofluorescence staining were used to assess the effects of AFAP1L1 on the progression of neoplasms and neovascular eye diseases in vivo. Transwell assay, wound scratch assay, tube forming assay, three-dimensional germination assay, and rhodamine-phalloidin staining were used to evaluate the impact of AFAP1L1 on human umbilical vein endothelial cells (HUVECs) function in vitro; Dual luciferase reporting, qRT-PCR and western blot were used to investigate the upstream and downstream mechanisms of pathological neovascularization mediated by AFAP1L1. Results Our investigation revealed that AFAP1L1 plays a crucial role in promoting the development of various tumors and demonstrates a strong correlation with endothelial cells. Targeted suppression of AFAP1L1 specifically in endothelial cells in vivo proves effective in inhibiting tumor formation and ocular pathological neovascularization. Mechanistically, AFAP1L1 functions as a hypoxia-related regulatory protein that can be activated by HIF-1α. In vitro experiments demonstrated that reducing AFAP1L1 levels can reverse hypoxia-induced excessive angiogenic capacity in HUVECs. The principal mechanism of angiogenesis inhibition entails the regulation of tip cell behavior through the YAP-DLL4-NOTCH axis. Conclusion In conclusion, AFAP1L1, a newly identified hypoxia-related regulatory protein, can be activated by HIF-1α. Inhibiting AFAP1L1 results in the inhibition of angiogenesis by suppressing the germination of endothelial tip cells through the YAP-DLL4-NOTCH axis. This presents a promising therapeutic target to halt the progression of tumors and neovascular eye disease.https://doi.org/10.1186/s12967-023-04503-xTumor angiogenesisOcular pathologic neovascularizationAFAP1L1HypoxiaHIF-1αVascular tip cell
spellingShingle Jun-Song Ren
Wen Bai
Jing-Juan Ding
Hui-Min Ge
Su-Yu Wang
Xi Chen
Qin Jiang
Hypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axis
Journal of Translational Medicine
Tumor angiogenesis
Ocular pathologic neovascularization
AFAP1L1
Hypoxia
HIF-1α
Vascular tip cell
title Hypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axis
title_full Hypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axis
title_fullStr Hypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axis
title_full_unstemmed Hypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axis
title_short Hypoxia-induced AFAP1L1 regulates pathological neovascularization via the YAP-DLL4-NOTCH axis
title_sort hypoxia induced afap1l1 regulates pathological neovascularization via the yap dll4 notch axis
topic Tumor angiogenesis
Ocular pathologic neovascularization
AFAP1L1
Hypoxia
HIF-1α
Vascular tip cell
url https://doi.org/10.1186/s12967-023-04503-x
work_keys_str_mv AT junsongren hypoxiainducedafap1l1regulatespathologicalneovascularizationviatheyapdll4notchaxis
AT wenbai hypoxiainducedafap1l1regulatespathologicalneovascularizationviatheyapdll4notchaxis
AT jingjuanding hypoxiainducedafap1l1regulatespathologicalneovascularizationviatheyapdll4notchaxis
AT huiminge hypoxiainducedafap1l1regulatespathologicalneovascularizationviatheyapdll4notchaxis
AT suyuwang hypoxiainducedafap1l1regulatespathologicalneovascularizationviatheyapdll4notchaxis
AT xichen hypoxiainducedafap1l1regulatespathologicalneovascularizationviatheyapdll4notchaxis
AT qinjiang hypoxiainducedafap1l1regulatespathologicalneovascularizationviatheyapdll4notchaxis