2009
DOI: 10.1074/jbc.m109.002378
|View full text |Cite
|
Sign up to set email alerts
|

Hyperglycemia Regulates RUNX2 Activation and Cellular Wound Healing through the Aldose Reductase Polyol Pathway

Abstract: Diabetes mellitus accelerates cardiovascular microangiopathies and atherosclerosis, which are a consequence of hyperglycemia. The aldose reductase (AR) polyol pathway contributes to these microvascular complications, but how it mediates vascular damage in response to hyperglycemia is less understood. The RUNX2 transcription factor, which is repressed in diabetic animals, promotes vascular endothelial cell (EC) migration, proliferation, and angiogenesis. Here we show that physiological levels of glucose (euglyc… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1
1

Citation Types

4
38
0

Year Published

2011
2011
2016
2016

Publication Types

Select...
7
1

Relationship

6
2

Authors

Journals

citations
Cited by 32 publications
(42 citation statements)
references
References 43 publications
(50 reference statements)
4
38
0
Order By: Relevance
“…HMVEC-Ls and HPAECs were studied in passages 2-7. Human bone marrow ECs (HBMEs) (31) and human umbilical vein retroviral telemorized ECs (Hrvts) were similarly cultured.…”
Section: Methodsmentioning
confidence: 99%
“…HMVEC-Ls and HPAECs were studied in passages 2-7. Human bone marrow ECs (HBMEs) (31) and human umbilical vein retroviral telemorized ECs (Hrvts) were similarly cultured.…”
Section: Methodsmentioning
confidence: 99%
“…Hyperglycemia (HG) elevates oxidative stress through the aldose reductase (AR) alternative glucose utilization pathway, which inhibits RUNX2 activity in bone marrow EC (HBME) without altering the levels of total RUNX2 protein (D’Souza et al, 2009). To determine whether this response was conserved in other cells, primary adipose (HAdEC), dermal (HMEC1), and umbilical vein (HRVT) ECs, as well as the classic glucose-responsive cell, the pancreatic β-cell (RINr) were treated with glucose after starvation to lower basal levels of RUNX2 DNA binding.…”
Section: Resultsmentioning
confidence: 99%
“…Recent findings from our laboratory revealed that RUNX2 is regulated by nutrients in the microenvironment and its DNA-binding activity is especially sensitive to glycemic status (D’Souza et al, 2009). HG inhibited, while anti-oxidants increased, RUNX2 activity and wound healing (D’Souza et al, 2009). Glucose activated RUNX2 DNA binding by increasing phosphorylation through the cdk pathway, which is less active in HG (Pierce et al, 2012).…”
Section: Introductionmentioning
confidence: 99%
“…In contrast, the quantitative D-ELISA is able to measure the interaction of RUNX2 with DNA-binding sequences corresponding to defined promoter elements in RUNX2 target genes. The use of an anti-RUNX2 antibody increased the specificity of the assay and distinguishes this assay from the traditional gel shift assay 5 . While the D-ELISA is an in vitro assay and cannot survey promoter occupancy in live cells, which is possible with ChIP assays, it can be used to quantitatively screen for compounds that inhibit the DNA-binding complexes.…”
Section: Incubation With Nuclear Extractmentioning
confidence: 99%
“…Use of an anti-RUNX2 antibody provides specificity to the assay and the lack of radiolabel distinguish this assay from the traditional gel shift assay 5 . Detection of binding complexes is possible with the use of a secondary antibody coupled to horseradish peroxidase (HRP), which converts an HRP substrate, tetramethyl benzidine (TMB) to a colored product for spectrophotometric analysis.…”
Section: Introductionmentioning
confidence: 99%