Abstract

Hypoxia inducible factors (HIFs) mediate angiogenesis via up-regulation of various pro-angiogenic factors (particularly VEGF) in response to hypoxia. Here, we report that hypoxia unexpectedly induced robust production of the pro-inflammatory factor TNFα by endothelial cells (ECs), suggesting an autocrine loop that in turn activated HIFs via an NF-κB-dependent process, resulting in production of VEGF and thereby promotion of angiogenesis. In contrast, low-density lipoprotein (LDL) prevented expression of HIFs in ECs exposed to either hypoxia or TNFα, while knockdown of either HIF-1α or HIF-2α strikingly attenuated hypoxia-induced production of VEGF by ECs as well as EC colony formation and tube formation. Significantly, LDL attenuated hypoxia-induced angiogenesis by disrupting the TNFα/NF-κB/HIF/VEGF signaling cascade via down-regulation of the TNF receptor TNF-R1, rather than TNFα itself, and multiple key components of both canonical and non-canonical NF-κB pathways. By doing so, LDL was able to either inhibit or down-regulate a wide spectrum of HIF-dependent pro-angiogenic downstream targets and signals. Together, these findings argue existence of a self-regulatory TNFα/NF-κB/HIF/VEGF signaling network in ECs, which mediates and fine-tones angiogenesis, at least in response to hypoxia. They also suggest that LDL impairs angiogenesis by disrupting this network, which might represent a novel mechanism underlying anti-angiogenic property of LDL.

Highlights

  • Hypoxia-driven angiogenesis represents one of the major adaptive responses to low oxygen supply, which is essential for maintenance of cellular metabolism, survival, and function, thereby representing a prominent feature of embryo development, ischemic injury recovery, tissue regeneration, inflammation, tumor growth, www.aging‐us.com etc. [1]

  • These results raise a possibility that a network involving four essential angiogenesis-driving signaling pathways including Hypoxia inducible factors (HIFs), TNF, VEGF, and NF-κB might be required for development of embryonic stem cells to endothelial cells (ECs), while it might be largely silenced after maturation of ECs no matter where they localize

  • A majority of genes involved in the HIF, TNF, VEGF, and NF-κB pathways were turned off once hESCs differentiated to mature ECs, while only some genes (e.g., HIF1A, TNFRSF1A, MAP3K14/ NIK) likely remained activated in human umbilical vein endothelial cells (HUVEC) when compared to HUAVCs

Read more

Summary

Introduction

Hypoxia-driven angiogenesis represents one of the major adaptive responses to low oxygen supply, which is essential for maintenance of cellular metabolism, survival, and function, thereby representing a prominent feature of embryo development, ischemic injury recovery, tissue regeneration, inflammation, tumor growth, www.aging‐us.com etc. [1]. It has been well established that as a major transcription factor, the HIF complex consisting of an oxygen-sensitive subunit (HIF-1α or HIF-2α) and a constitutively expressed HIF1β plays an essential role in driving angiogenesis under various physiological and pathological conditions [4,5]. In the latter case, the significance of HIF-mediated angiogenesis (via VEGF) has been well documented in a wide variety of disease types, especially aging-related diseases such as atherosclerotic (e.g., coronary artery disease, ischemic stroke) [5] and degenerative disorders (e.g., neurodegeneration [6] and macular degeneration [7]). In diverse types of cells (especially ECs), downstream genes of HIF are involved in vascular remodeling, inflammation, cell survival and proliferation, apoptosis, autophagy, cell migration and invasion, DNA damage response, extracellular matrix metabolism, as well as glucose metabolism (e.g., glucose uptake and glycolysis) [8,9]

Methods
Results
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