Abstract

BackgroundEndothelial cell (EC) injury accelerates the progression of diabetic macrovascular complications. Hypoxia is an important cause of EC injury. Hypoxia-inducible factor-1 alpha (HIF-1α) is an important hypoxia regulatory protein. Our previous studies showed that high-glucose and hypoxic conditions could upregulate HIF-1α expression and enhance EC inflammatory injury, independently of the nuclear factor kappa-B (NF-κB) pathway. However, it is not clear whether HIF-1α plays a role in vascular disease through epigenetic-related mechanisms.MethodsWe conducted gene expression analysis and molecular mechanistic studies in human umbilical vein endothelial cells (HUVECs) induced by hyperglycemia and hypoxia using RNA sequencing (RNA-seq) and small interfering HIF-1α (si-HIF-1α). We determined HIF-1α and Jumonji domain-containing protein 1 A (JMJD1A) expression by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot, analyzed inflammatory protein secretion in the cell supernatant by enzymelinked immunosorbent assay (ELISA), and assessed protein interaction between HIF-1α and JMJD1A by chromatin immunoprecipitation (Ch-IP). We used the Cell Counting Kit8 (CCK-8) assay to analyze cell viability, and assessed oxidative stress indicators by using a detection kit and flow cytometry.ResultsHigh glucose and hypoxia up-regulated HIF-1α expression, and down-regulated HIF-1α decreased the level of inflammation and oxidative stress in HUVECs. To determine the downstream pathways, we observed histone demethylases genes and related pathway by RNA-sEq. Among these, JMJD1A was the most upregulated gene in histone demethylases. Moreover, we observed that HIF-1α bound to the promoter of JMJD1A, and the ameliorative effects of si-HIF-1α on oxidative stress and inflammatory cytokines in high-glucose and hypoxia-induced HUVECs were reversed by JMJD1A overexpression. Furthermore, knockdown of JMJD1A decreased inflammatory and oxidative stress injury. To determine the JMJD1A-related factors, we conducted gene expression analysis on JMJD1A-knockdown HUVECs. We observed that downregulation of inflammation and the oxidative stress pathway were enriched and FOS and FOSB might be important protective transcription factors.ConclusionsThese findings provide novel evidence that the HIF-1α/JMJD1A signaling pathway is involved in inflammation and oxidative stress in HUVECs induced by high glucose and hypoxia. Also, this pathway might act as a novel regulator of oxidative stress and inflammatory-related events in response to diabetic vascular injury and thus contribute to the pathological progression of diabetes and vascular disease.

Highlights

  • Endothelial cell (EC) injury accelerates the progression of diabetic macrovascular complications

  • High glucose decreased cell viability and increased inflammation and reactive oxygen species (ROS) production in human umbilical vein endothelial cells (HUVECs) HUVECs exposed to different concentrations of high glucose (25 and 30 mM) exhibited lower cell survival, with 53.77 and 51.69% of cells surviving relative to normal conditions for 24 h and 49.29 and 52.1% for 48 h, respectively, showing the significantly cytotoxic effect of high glucose (p < 0.05, n = 3) (Fig. 1a and Additional file 1: Figure S1)

  • As far as we know, our study is the first to present the following findings: (i) Hypoxia-inducible factor-1α (HIF-1α)/ Jumonji domain-containing (JMJD1A) expression was induced by exposure to high-glucose and hypoxic conditions in a time-dependent manner and was positively associated with inflammatory and oxidative stress injury in HUVECs; and (ii) downregulation of HIF-1α/JMJD1A inhibited the excessive production of inflammatory and oxidative stress of HUVECs induced by high glucose and hypoxia

Read more

Summary

Introduction

Endothelial cell (EC) injury accelerates the progression of diabetic macrovascular complications. Our previous studies showed that high-glucose and hypoxic conditions could upregulate HIF-1α expression and enhance EC inflammatory injury, independently of the nuclear factor kappa-B (NF-κB) pathway. It is not clear whether HIF-1α plays a role in vascular disease through epigenetic-related mechanisms. Studies have confirmed that diabetic patients suffer from 1.45 to 2.99 times the risk of atherosclerosis in non-diabetic patients [6], and the incidence of endothelial cell (EC)-related complications is about 2–4 times higher than in individuals without diabetes [7] It is important for patients with diabetic macrovascular complications to find effective EC protection strategies [8, 9]. The molecular mechanisms underlying DM-related macrovascular complication require clarification

Objectives
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